1
|
Wang J, Du Y, Peng Y, Deng Y, Ge Y, Liu Z, Lv J, Hu G, Zhao Z, Li Y. Prevalence and network structure of depression, anxiety and adverse doctor-patient relationship risks among patients with physical diseases: A cross-sectional study. J Affect Disord 2025; 376:122-130. [PMID: 39884365 DOI: 10.1016/j.jad.2025.01.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Mental health issues among patients with physical diseases are increasingly common. This study investigated the prevalence of depression, anxiety, and adverse doctor-patient relationship risks (ADRR) among patients with physical diseases, and the central and bridge symptoms of this network structure. METHODS A total of 14,344 patients with physical diseases enrolled in this survey. The Patient Health Questionnaire-9 (PHQ-9), Generalized Anxiety Disorder-7 (GAD-7), and Psychological Safety Questionnaire were used to evaluate anxiety, depression, and ADRR. The "qgraph" package in R 4.4.3 was used to construct a network model to identify central and bridge symptoms. RESULTS The prevalence rates of depression, anxiety, and ADRR were found to be 9.52 % (95 % confidence interval (CI): 9.04-10.00 %), 19.35 % (95 % CI: 18.71-20.00 %), and 4.29 % (95 % CI: 3.96-4.62 %), respectively. Within the network structure, the central symptoms identified were 'Sad mood,' 'Restlessness,' and 'Excessive worry,' which also served as the bridge symptoms. The flow network analysis revealed that ADRR exhibited the strongest associations with 'Anhedonia', 'Restlessness', and 'Suicidal ideation'. Additionally, 'Suicidal ideation' shows strongest correlations with 'Guilt', 'Concentration', and 'Restlessness'. LIMITATION The generalizability of the study's findings is constrained, as the sample consisted exclusively of inpatients, potentially limiting applicability to non-hospitalized individuals with physical illnesses. CONCLUSION This study provides novel insights into the comorbidity of depression, anxiety, and ADRR at the symptom level in patients with physical diseases through the application of network analysis. The identification of bridge symptoms highlights potential targets for interventions aimed at addressing the comorbidity among these disorders.
Collapse
Affiliation(s)
- Jianqiang Wang
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China; College of Education, Hebei Normal University, Shijiazhuang, China
| | - Yuru Du
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China; Hebei Key Laboratory of Early Life Health Promotion, Hebei Medical University, Shijiazhuang, China
| | - Yuhan Peng
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China
| | - Yishan Deng
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China
| | - Yiran Ge
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China
| | - Zheng Liu
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China
| | - Jing Lv
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China; College of Education, Hebei Normal University, Shijiazhuang, China
| | - Gengdan Hu
- Department of Psychology, School of Humanities, Tongji University, Shanghai, China; Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Zengren Zhao
- Department of General Surgery, The First Hospital of Hebei Medical University, Hebei, China
| | - Youdong Li
- Clinical Mental Health Department, The First Hospital of Hebei Medical University, Hebei, China; Hebei Key Laboratory of Early Life Health Promotion, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
2
|
Fries LE, Dharma S, Chakravarti A, Chatterjee S. Variability in proliferative and migratory defects in Hirschsprung disease-associated RET pathogenic variants. Am J Hum Genet 2025; 112:863-875. [PMID: 40010351 DOI: 10.1016/j.ajhg.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
Hirschsprung disease (HSCR) exhibits extensive genetic heterogeneity, with 72% of cases involving pathogenic variants in 10 genes forming a gene regulatory network (GRN) essential for enteric nervous system (ENS) development. The receptor tyrosine kinase gene RET is the most significant contributor, implicated in 12%-50% of individuals depending on the phenotype. RET plays a critical role in ENS precursor proliferation and migration, and defects in these processes lead to HSCR. However, the functional impact of RET pathogenic variants and their mechanisms of disease remain poorly understood. To address this, we investigated proliferative and migratory phenotypes in a RET-dependent neural crest-derived cell line harboring one of five missense (c.166C>A [p.Leu56Met]; c.532G>C [p.Glu178Gln]; c.2372A>T [p.Tyr791Phe]; c.2765C>A [p.Ser922Tyr]; or c.2994T>A [p.Phe998Leu]) or three nonsense (c.612C>A, c.2308C>T, or c.2943C>G) heterozygous pathogenic RET variants. Using cDNA- and CRISPR-based prime reverse insertion mechanism engineering (PRIME) editing coupled with quantitative proliferation and migration assays, we observed significant losses in proliferation and migration in three missense (c.612C>A [p.Tyr204∗]; c.2308C>T [p.Arg770∗]; and c.2943C>G [p.Tyr981∗]) and all nonsense variants. Notably, the c.2372A>T (p.Tyr791Phe) missense variant, whose pathogenicity has been debated, appears benign. Importantly, the severity of migration loss did not consistently correlate with proliferation defects, and the phenotypic severity of nonsense variants was independent of their position within the RET protein. This study highlights the necessity of targeted functional assays to accurately assess the pathogenicity of HSCR-associated variants rather than relying solely on bioinformatics predictions, which could be refined by incorporating functional data.
Collapse
Affiliation(s)
- Lauren E Fries
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sree Dharma
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Aravinda Chakravarti
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| | - Sumantra Chatterjee
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
3
|
Zhong Y, So MT, Ma Z, Zhang D, Wang Y, Xiong Z, Fadista J, Song YQ, Cheah KSE, Alves MM, Borrego S, Ceccherini I, Pakarinen MP, Feenstra B, Lui VCH, Garcia-Barcelo MM, Sham PC, Tam PKH, Tang CSM. Multi-ancestry genome-wide association meta-analysis identifies novel associations and informs genetic risk prediction for Hirschsprung disease. EBioMedicine 2025; 115:105680. [PMID: 40184909 DOI: 10.1016/j.ebiom.2025.105680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Hirschsprung disease (HSCR) is a rare, congenital disease characterized by the absence of enteric ganglia in the hindgut. Common genetic variation contributes substantially to the heritability of the disease yet only three HSCR-associated loci were identified from genome-wide association studies (GWAS) thus far. METHODS We performed the largest multi-ancestry meta-analysis of GWAS to date, totalling 1250 HSCR cases and 7140 controls. Prioritized candidate genes were further characterized using single-cell transcriptomic data of developing human and mouse gut for their roles in development of enteric nervous system (ENS). Functional characterisation using human cells and zebrafish models was performed. Global and ancestry-matched polygenic risk score (PRS) models were derived and evaluated for predicting risk of HSCR. FINDINGS We identified four HSCR-susceptibility loci, with three loci (JAG1, HAND2 and ZNF25) reaching genome-wide significance and one putative locus (UNC5C) prioritized by functional relevance. Spatiotemporal analysis revealed hotspots of gene dysregulation during ENS development. Functional analyses further demonstrated that knockdown of the candidate genes impaired cell migration and zebrafish knockouts displayed abnormal ENS development. We also demonstrated comparable performance for a PRS model derived from multi-ancestry meta-analysis to those of ancestry-matched PRS models, supporting its potential clinical application in risk prediction of HSCR across populations. INTERPRETATION Overall, the meta-analysis implicated novel genes, pathways and spatiotemporal developmental hotspots in the genetic aetiology of HSCR. Development of a PRS universally applicable irrespective of ancestries may leverage its clinical utility in risk prediction. FUNDING The full list of funding bodies can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Yuanxin Zhong
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Man-Ting So
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Zuyi Ma
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Detao Zhang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Yanbing Wang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Zewei Xiong
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - João Fadista
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - You-Qiang Song
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Kathryn Song-Eng Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Centre, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Salud Borrego
- Department of Genetics, Reproduction and Fetal Medicine, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocio/CSIC/University of Seville, Seville, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | | | - Mikko P Pakarinen
- Section of Pediatric Surgery, Helsinki University Hospital and University of Helsinki, Finland; Pediatric Liver and Gut Research Group, University of Helsinki, Finland
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Vincent Chi-Hang Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China; Dr Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong Special Administrative Region of China
| | - Maria-Merce Garcia-Barcelo
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Pak Chung Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Paul Kwong-Hang Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China; Faculty of Medicine, Macau University of Science and Technology, Macao, China.
| | - Clara Sze-Man Tang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China; Dr Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong Special Administrative Region of China.
| |
Collapse
|
4
|
Horváth E, Derzsi Z, Löckli E, Molnár GB, Bara Z, Kovács E, Gozar H. Histopathologic Concerns and Diagnostic Challenges in Hirschsprung's Disease: An Eastern European Single-Center Observational Study. Life (Basel) 2025; 15:329. [PMID: 40141676 PMCID: PMC11943527 DOI: 10.3390/life15030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND We proposed a comprehensive clinicopathological study involving the characterization of the study cohort and a comparative analysis of biopsies and surgical specimens from patients with Hirschsprung's disease. The study was complemented by the diagnostic value of calretinin, CD56, and S-100 immunohistochemistry. METHODS Descriptive statistical analysis of diagnostic variables in the group of biopsy specimens (n = 32) and bowel resection specimens (n = 16) was performed. The pattern of calretinin and CD56 expression in Meissner's plexus elements was analyzed and the maximum thicknesses of the nerve fibers were measured using morphometry with S100-immunostained sections. CONCLUSIONS Coupled calretinin-CD56 immunohistochemistry is useful in diagnosing ganglion cell paucity biopsies or specimens with incomplete submucosa. In cases where there are no ganglion cells but there are calretinin-positive nerve fibrils in the lamina propria without nerve trunk (NT) hypertrophy, re-biopsy is the best solution. The significant differences in NT size between biopsies and surgical specimens highlight the importance of assessing NT diameter in all tissue samples examined.
Collapse
Affiliation(s)
- Emőke Horváth
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Pathology Service, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Zoltán Derzsi
- Department of Pediatric Surgery and Orthopedics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (Z.B.); (H.G.)
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Eliza Löckli
- George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Gyopár-Beáta Molnár
- Pathology Service, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Zsolt Bara
- Department of Pediatric Surgery and Orthopedics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (Z.B.); (H.G.)
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Evelyn Kovács
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Horea Gozar
- Department of Pediatric Surgery and Orthopedics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (Z.B.); (H.G.)
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| |
Collapse
|
5
|
Çoşkun N, Metin M. Scientific evolution from the definition of Hirschsprung disease to the present: a bibliometric analysis (1980-2023). Pediatr Res 2025:10.1038/s41390-025-03927-z. [PMID: 39979585 DOI: 10.1038/s41390-025-03927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND The primary objective of this study is to define the global productivity of Hirschsprung disease (HSCR), identify influential studies, determine research directions focused on both historical and contemporary perspectives. METHODS The study obtained 2816 articles published between 1980 and 2023 related to HSCR from the Web of Science database, and comprehensive bibliometric analysis were conducted. RESULTS The top three most productive countries were the USA (n = 1283), China (n = 1167), and Japan (n = 587). The most productive institution was Université Paris Cité (n = 149), and the most productive author was Prem Puri (n = 99). The most frequently used keywords in the articles were enterocolitis (n = 191), enteric nervous system (n = 136), and transanal endorectal pull-through (n = 129). CONCLUSION Academic interest in HSCR began in 1887-1888, significantly increased in 1948 and 1964, and reached its highest peak in 1994. The findings indicate that key topics such as enterocolitis, enteric nervous system, surgical techniques, and genetic factors have been prominent in HSCR research. Previous studies, which often focused on molecular and genetic mechanisms, have shifted towards clinical and surgical applications in the last decade. Factor analysis reveals the complexity and diversity of HSCR research, with various topics examined, including clinical features, surgical treatments, pathological findings, and genetic bases. IMPACT STATEMENT This study presents a comprehensive bibliometric analysis of global research trends and future directions in Hirschsprung disease, revealing significant changes and developments in the study of the disease. The study provides an important contribution to the existing literature by detailing the historical development, main research topics, and thematic evolution of Hirschsprung disease research. The findings indicate that future research focus in Hirschsprung disease may increasingly prioritize innovative approaches, such as stem cell therapy, alongside clinical and surgical advancements. These advancements have the potential to enhance patient quality of life and guide future research strategies.
Collapse
Affiliation(s)
- Nurcan Çoşkun
- Department of Pediatric Surgery, Hitit University Erol Olçok Training and Research Hospital, Hitit University, Çorum, Turkey.
| | - Mehmet Metin
- Faculty of Medicine, Department of Pediatric Surgery, Hitit University, Çorum, Turkey
| |
Collapse
|
6
|
Tjaden NEB, Liou MJ, Sax SE, Lassoued N, Lou M, Schneider S, Beigel K, Eisenberg JD, Loeffler E, Anderson SE, Yan G, Litichevskiy L, Dohnalová L, Zhu Y, Jin DMJC, Raab J, Furth EE, Thompson Z, Rubenstein RC, Pilon N, Thaiss CA, Heuckeroth RO. Dietary manipulation of intestinal microbes prolongs survival in a mouse model of Hirschsprung disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637436. [PMID: 39990395 PMCID: PMC11844371 DOI: 10.1101/2025.02.10.637436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Enterocolitis is a common and potentially deadly manifestation of Hirschsprung disease (HSCR) but disease mechanisms remain poorly defined. Unexpectedly, we discovered that diet can dramatically affect the lifespan of a HSCR mouse model ( Piebald lethal , sl/sl ) where affected animals die from HAEC complications. In the sl/sl model, diet alters gut microbes and metabolites, leading to changes in colon epithelial gene expression and epithelial oxygen levels known to influence colitis severity. Our findings demonstrate unrecognized similarity between HAEC and other types of colitis and suggest dietary manipulation could be a valuable therapeutic strategy for people with HSCR. Abstract Hirschsprung disease (HSCR) is a birth defect where enteric nervous system (ENS) is absent from distal bowel. Bowel lacking ENS fails to relax, causing partial obstruction. Affected children often have "Hirschsprung disease associated enterocolitis" (HAEC), which predisposes to sepsis. We discovered survival of Piebald lethal ( sl/sl ) mice, a well-established HSCR model with HAEC, is markedly altered by two distinct standard chow diets. A "Protective" diet increased fecal butyrate/isobutyrate and enhanced production of gut epithelial antimicrobial peptides in proximal colon. In contrast, "Detrimental" diet-fed sl/sl had abnormal appearing distal colon epithelium mitochondria, reduced epithelial mRNA involved in oxidative phosphorylation, and elevated epithelial oxygen that fostered growth of inflammation-associated Enterobacteriaceae . Accordingly, selective depletion of Enterobacteriaceae with sodium tungstate prolonged sl/sl survival. Our results provide the first strong evidence that diet modifies survival in a HSCR mouse model, without altering length of distal colon lacking ENS. Highlights Two different standard mouse diets alter survival in the Piebald lethal ( sl/sl ) mouse model of Hirschsprung disease, without impacting extent of distal colon aganglionosis (the region lacking ENS). Piebald lethal mice fed the "Detrimental" diet had many changes in colon epithelial transcriptome including decreased mRNA for antimicrobial peptides and genes involved in oxidative phosphorylation. Detrimental diet fed sl/sl also had aberrant-appearing mitochondria in distal colon epithelium, with elevated epithelial oxygen that drives lethal Enterobacteriaceae overgrowth via aerobic respiration. Elimination of Enterobacteriaceae with antibiotics or sodium tungstate improves survival of Piebald lethal fed the "Detrimental diet". Graphical abstract
Collapse
|
7
|
Butler Tjaden NE, Shannon SR, Seidel CW, Childers M, Aoto K, Sandell LL, Trainor PA. Rdh10-mediated Retinoic Acid Signaling Regulates the Neural Crest Cell Microenvironment During ENS Formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634504. [PMID: 39896510 PMCID: PMC11785139 DOI: 10.1101/2025.01.23.634504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The enteric nervous system (ENS) is formed from vagal neural crest cells (NCC), which generate most of the neurons and glia that regulate gastrointestinal function. Defects in the migration or differentiation of NCC in the gut can result in gastrointestinal disorders such as Hirschsprung disease (HSCR). Although mutations in many genes have been associated with the etiology of HSCR, a significant proportion of affected individuals have an undetermined genetic diagnosis. Therefore, it's important to identify new genes, modifiers and environmental factors that regulate ENS development and disease. Rdh10 catalyzes the first oxidative step in the metabolism of vitamin A to its active metabolite, RA, and is therefore a central regulator of vitamin A metabolism and retinoic acid (RA) synthesis during embryogenesis. We discovered that retinol dehydrogenase 10 (Rdh10) loss-of-function mouse embryos exhibit intestinal aganglionosis, characteristic of HSCR. Vagal NCC form and migrate in Rdh10 mutant embryos but fail to invade the foregut. Rdh10 is highly expressed in the mesenchyme surrounding the entrance to the foregut and is essential between E7.5-E9.5 for NCC invasion into the gut. Comparative RNA-sequencing revealed downregulation of the Ret-Gdnf-Gfrα1 gene signaling network in Rdh10 mutants, which is critical for vagal NCC chemotaxis. Furthermore, the composition of the extracellular matrix through which NCC migrate is also altered, in part by increased collagen deposition. Collectively this restricts NCC entry into the gut, demonstrating that Rdh10-mediated vitamin A metabolism and RA signaling pleiotropically regulates the NCC microenvironment during ENS formation and in the pathogenesis of intestinal aganglionosis.
Collapse
Affiliation(s)
- Naomi E. Butler Tjaden
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Gastroenterology, Hepatology & Nutrition, Children’s Hospital of Philadelphia, Philadelphia PA 19104
| | - Stephen R. Shannon
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | - Melissa Childers
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu City, Shizuoka, Japan 431-3192
| | - Lisa L. Sandell
- University of Louisville, Department of Oral Immunology and Infectious Diseases, Louisville, KY, 40201, USA
| | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Zhang Q, He Y, Rao D, He R, Yu L, Sun Y, Lai Y, Shi Z, Peng L, Zhang Z, Xu S. Discovery of an Efficacious RET PROTAC Degrader with Enhanced Antiproliferative Activity against Resistant Cancer Cells Harboring RET Solvent-Front Mutations. J Med Chem 2025; 68:753-775. [PMID: 39731581 DOI: 10.1021/acs.jmedchem.4c02692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Rearranged during transfection (RET) kinase is a validated therapeutic target for various cancers characterized by RET alterations. Although two selective RET inhibitors, selpercatinib and pralsetinib, have been approved by the FDA, acquired resistance through solvent-front mutations has been identified rapidly. Developing proteolysis targeting chimera (PROTAC) targeting RET mutations offers a promising strategy to combat drug resistance. Herein, we describe the design, synthesis, and evaluation of a series of RET PROTAC degraders. The representative compound QZ2135 (20) effectively degraded RET kinase and its resistant mutants, such as V804M and G810C/R. It also exhibited superior antiproliferative activity against Ba/F3 cells stably expressing oncogenic fusions of RET with solvent-front mutants, including G810C/R/S, compared to its parental inhibitor. Notably, QZ2135 demonstrated in vivo antitumor efficacy in a Ba/F3-KIF5B-RET-G810C xenograft mouse model. Together, this study provides a potential alternative strategy for overcoming acquired resistance to RET inhibitors mediated by solvent-front mutations.
Collapse
Affiliation(s)
- Qian Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yingqi He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Danni Rao
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuanhui Lai
- Department of Thyroid and Breast Surgery, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510310, China
| | - Zihan Shi
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lijie Peng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhang Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, School of Pharmacy, Jinan University, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
- Department of Thyroid and Breast Surgery, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510310, China
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Wattchow DA, Brookes SJH, Spencer NJ, De Giorgio R, Costa M, Dinning PG. Gut Neuropathies and Intestinal Motility Disorders. Neurogastroenterol Motil 2025:e14995. [PMID: 39777822 DOI: 10.1111/nmo.14995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 11/26/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The enteric nervous system plays a key role in the coordination of gastrointestinal motility together with sympathetic, parasympathetic, and extrinsic sensory pathways. In some cases, abnormalities in neural activity in these pathways contribute to disorders of gut motility. Where this is associated with damage or death of enteric neurons, usually detected by microscopy, this is considered a gut neuropathy. PURPOSE This review summarizes recent advances in the identification of neuropathies in a range of gastrointestinal motility disorders.
Collapse
Affiliation(s)
- David A Wattchow
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Department of Surgery, Flinders Medical Centre, Adelaide, South Australia, Australia
- Department of Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Simon J H Brookes
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Marcello Costa
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Department of Surgery, Flinders Medical Centre, Adelaide, South Australia, Australia
- Department of Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|
10
|
Fu M, Berk-Rauch HE, Chatterjee S, Chakravarti A. The Role of de novo and Ultra-Rare Variants in Hirschsprung Disease (HSCR): Extended Gene Discovery for Risk Profiling of Patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.07.25320162. [PMID: 39830246 PMCID: PMC11741498 DOI: 10.1101/2025.01.07.25320162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Hirschsprung disease (HSCR) is a rare neurodevelopmental disorder caused by disrupted migration and proliferation of enteric neural crest cells during enteric nervous system development. Genetic studies suggest a complex etiology involving both rare and common variants, but the contribution of ultra-rare pathogenic variants (PAs) remains poorly understood. Methods We perform whole-exome sequencing (WES) on 301 HSCR probands and 109 family trios, employing advanced statistical methods and gene prioritization strategies to identify genes carrying de novo and ultra-rare coding pathogenic variants. Multiple study designs, including case-control, de novo mutation analysis and joint test, are used to detect associated genes. Candidate genes are further prioritized based on their biological and functional relevance to disease associated tissues and onset period (i.e., human embryonic colon). Results We identify 19 risk genes enriched with ultra-rare coding pathogenic variants in HSCR probands, including four known genes (RET, EDNRB, ZEB2, SOX10) and 15 novel candidates (e.g., COLQ, NES, FAT3) functioning in neural proliferation and neuromuscular synaptic development. These genes account for 17.5% of the population-attributable risk (PAR), with novel candidates contributing 6.5%. Notably, a positive correlation between pathogenic mutational burden and disease severity is observed. Female cases exhibit at least 42% higher ultra-rare pathogenic variant burden than males (P = 0.05). Conclusions This first-ever genome-wide screen of ultra-rare variants in a large, phenotypically diverse HSCR cohort highlights the substantial contribution of ultra-rare pathogenic variants to the disease risk and phenotypic variability. These findings enhance our understanding of the genetic architecture of HSCR and provide potential targets for genetic screening and personalized interventions.
Collapse
Affiliation(s)
- Mingzhou Fu
- Center for Human Genetics and Genomics, New York University
Grossman School of Medicine, New York, NY, 10016
- Department of Population Health, New York University Grossman
School of Medicine, New York, NY, 10016
| | - Hanna E Berk-Rauch
- Center for Human Genetics and Genomics, New York University
Grossman School of Medicine, New York, NY, 10016
| | - Sumantra Chatterjee
- Center for Human Genetics and Genomics, New York University
Grossman School of Medicine, New York, NY, 10016
- Department of Neuroscience and Physiology, New York University
Grossman School of Medicine, New York, NY, 10016
| | - Aravinda Chakravarti
- Center for Human Genetics and Genomics, New York University
Grossman School of Medicine, New York, NY, 10016
- Department of Neuroscience and Physiology, New York University
Grossman School of Medicine, New York, NY, 10016
| |
Collapse
|
11
|
Windster JD, Kakiailatu NJM, Kuil LE, Antanaviciute A, Sacchetti A, MacKenzie KC, Peulen-Zink J, Kan TW, Bindels E, de Pater E, Doukas M, van den Bosch TPP, Yousefi S, Barakat TS, Meeussen CJHM, Sloots PCEJ, Wijnen RMH, Parikh K, Boesmans W, Melotte V, Hofstra RMW, Simmons A, Alves MM. Human Enteric Glia Diversity in Health and Disease: New Avenues for the Treatment of Hirschsprung Disease. Gastroenterology 2024:S0016-5085(24)05803-7. [PMID: 39725172 DOI: 10.1053/j.gastro.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/16/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND & AIMS The enteric nervous system (ENS), which is composed of neurons and glia, regulates intestinal motility. Hirschsprung disease (HSCR) results from defects in ENS formation; however, although neuronal aspects have been studied extensively, enteric glia remain disregarded. This study aimed to explore enteric glia diversity in health and disease. METHODS Full-thickness intestinal resection material from pediatric controls and patients with HSCR was collected, dissociated, and enriched for the ENS population through fluorescence-activated cell sorting. Single-cell RNA sequencing was performed to uncover the transcriptomic diversity of the ENS in controls and HSCR patients, as well as in wild-type and ret mutant zebrafish. Immunofluorescence and fluorescence in situ hybridization confirmed the presence of distinct subtypes. RESULTS Two major enteric glial classes emerged in the pediatric intestine: Schwann-like enteric glia, which are reminiscent of Schwann cells, and enteric glia expressing classical glial markers. Comparative analysis with previously published datasets confirmed our classification and revealed that although classical enteric glia are predominant prenatally, Schwann-like enteric glia become more abundant postnatally. In HSCR, ganglionic segments mirrored controls and aganglionic segments featured only Schwann-like enteric glia. Leveraging the regenerative potential of Schwann cells, we explored therapeutic options using a ret mutant zebrafish. Prucalopride, a serotonin-receptor (5-HT) agonist, induced neurogenesis partially rescuing the HSCR phenotype in ret+/- mutants. CONCLUSIONS Two major enteric glial classes were identified in the pediatric intestine, highlighting the significant postnatal contribution of Schwann-like enteric glia to glial heterogeneity. Crucially, these glial subtypes persist in aganglionic segments of patients with HSCR, offering a new target for their treatment using 5-HT agonists.
Collapse
Affiliation(s)
- Jonathan D Windster
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Naomi J M Kakiailatu
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Agne Antanaviciute
- MRC Translational Immune Discovery Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom; MRC WIMM Centre for Computational Biology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Andrea Sacchetti
- Department of Pathology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Katherine C MacKenzie
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Joke Peulen-Zink
- Department of Hematology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tsung W Kan
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eric Bindels
- Department of Hematology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Emma de Pater
- Department of Hematology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thierry P P van den Bosch
- Department of Pathology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Soheil Yousefi
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Tahsin-Stefan Barakat
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Conny J H M Meeussen
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Pim C E J Sloots
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Rene M H Wijnen
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Kaushal Parikh
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Werend Boesmans
- Biomedical Research Institute, Hasselt University, Hasselt, Belgium; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Veerle Melotte
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Alison Simmons
- MRC WIMM Centre for Computational Biology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands.
| |
Collapse
|
12
|
Ghandour F, Green KD, Jain E, Bajpai P, Patel CR, Manne U, Diffalha SA. Novel ganglion cell marker B3GNT6: A step forward in Hirschsprung's disease diagnosis. Pathol Res Pract 2024; 266:155780. [PMID: 39729957 DOI: 10.1016/j.prp.2024.155780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Hirschsprung's (HSCR) disease, also known as aganglionic megacolon, or congenital intestinal aganglionosis affects roughly 1 out of every 5000 newborns. It is a birth defect characterized by the partial or complete loss of ganglion cells in the myenteric and submucosal plexus of the distal intestine which leads to ineffective peristalsis, constipation, and obstruction. Clinical assessment and radiological observations might imply HSCR disease, but definitive diagnosis requires biopsy interpretation and confirmation of ganglion cell loss. The difficulty in identifying immature ganglion cells added to the variability in interpreting immunohistochemical markers of ganglion cells warrants the search for new markers. Our recent research identified Beta-1,3-N-acetylglucosaminyltransferase (B3GNT6) as a potential candidate, as it consistently stains the cytoplasm of ganglion cells. To evaluate its utility, we conducted a preliminary assessment of B3GNT6 expression in nineteen gastrointestinal tissue samples and observed cytoplasmic staining in ganglion cells across all samples. This consistent staining pattern suggests B3GNT6 could serve as a reliable marker for diagnosing Hirschsprung's disease. This article serves as a preliminary evaluation of B3GNT6 as a ganglion cell immunohistochemical marker, highlighting its potential significance while acknowledging the need for further validation in larger, more diverse cohorts.
Collapse
Affiliation(s)
- Fatme Ghandour
- University of Alabama at Birmingham, Department of Pathology, United States.
| | - Kesley D Green
- University of Alabama at Birmingham, Department of Pathology, United States.
| | - Ekta Jain
- University of Alabama at Birmingham, Department of Pathology, United States.
| | - Prachi Bajpai
- University of Alabama at Birmingham, Department of Pathology, United States.
| | | | - Upender Manne
- University of Alabama at Birmingham, Department of Pathology, United States.
| | - Sameer Al Diffalha
- University of Alabama at Birmingham, Department of Pathology, United States.
| |
Collapse
|
13
|
Magel V, Blum J, Dolde X, Leisner H, Grillberger K, Khalidi H, Gardner I, Ecker GF, Pallocca G, Dreser N, Leist M. Inhibition of Neural Crest Cell Migration by Strobilurin Fungicides and Other Mitochondrial Toxicants. Cells 2024; 13:2057. [PMID: 39768149 PMCID: PMC11674305 DOI: 10.3390/cells13242057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Cell-based test methods with a phenotypic readout are frequently used for toxicity screening. However, guidance on how to validate the hits and how to integrate this information with other data for purposes of risk assessment is missing. We present here such a procedure and exemplify it with a case study on neural crest cell (NCC)-based developmental toxicity of picoxystrobin. A library of potential environmental toxicants was screened in the UKN2 assay, which simultaneously measures migration and cytotoxicity in NCC. Several strobilurin fungicides, known as inhibitors of the mitochondrial respiratory chain complex III, emerged as specific hits. From these, picoxystrobin was chosen to exemplify a roadmap leading from cell-based testing towards toxicological predictions. Following a stringent confirmatory testing, an adverse outcome pathway was developed to provide a testable toxicity hypothesis. Mechanistic studies showed that the oxygen consumption rate was inhibited at sub-µM picoxystrobin concentrations after a 24 h pre-exposure. Migration was inhibited in the 100 nM range, under assay conditions forcing cells to rely on mitochondria. Biokinetic modeling was used to predict intracellular concentrations. Assuming an oral intake of picoxystrobin, consistent with the acceptable daily intake level, physiologically based kinetic modeling suggested that brain concentrations of 0.1-1 µM may be reached. Using this broad array of hazard and toxicokinetics data, we calculated a margin of exposure ≥ 80 between the lowest in vitro point of departure and the highest predicted tissue concentration. Thus, our study exemplifies a hit follow-up strategy and contributes to paving the way to next-generation risk assessment.
Collapse
Affiliation(s)
- Viktoria Magel
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Jonathan Blum
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Xenia Dolde
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Heidrun Leisner
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
| | - Karin Grillberger
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Hiba Khalidi
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Iain Gardner
- Certara Predictive Technologies, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, UK
| | - Gerhard F. Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Giorgia Pallocca
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Nadine Dreser
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78464 Konstanz, Germany
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| |
Collapse
|
14
|
Olsbø S, Kiserud SG, Hermansen Å, Hamilton Larsen M, Bjørnland K. Health literacy in parents of children with Hirschsprung disease: a novel study. Pediatr Surg Int 2024; 41:21. [PMID: 39633025 PMCID: PMC11618141 DOI: 10.1007/s00383-024-05917-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE To explore health literacy (HL) among parents of children with Hirschsprung disease (HD). METHODS Norwegian-speaking parents of children under 16 who underwent HD surgery at a tertiary center were surveyed using the Health Literacy Questionnaire-Parent, electronic Health Literacy Scale, General Self-efficacy Scale, and a study-specific questionnaire. Demographics were collected and ethical approval was obtained. RESULTS Among 132 parents (77 mothers) of 91 children (median age 8 years), high HL scores appeared in the domains "understanding health information" and "active engagement", with lower scores in "provider support", "health information appraisal", and "social support". Higher HL correlated with parents aged over 40 and higher education. Lower scores were seen with non-exclusive Norwegian use at home and not living with the child's other parent. High electronic HL scores were common (mean 3.6, maximum score 5). 69% had high self-efficacy scores (score > 2, maximum score 4). Self-efficacy correlated strongly with higher HL scores. CONCLUSION Parents of children with HD feel healthcare providers lack understanding of their child's challenges, experience limited social support and struggle with interpreting health information. We suggest targeted HL interventions for young, lower-educated, non-cohabitating parents and those not primarily speaking the official language at home.
Collapse
Affiliation(s)
- Signe Olsbø
- Department of Gastrointestinal and Pediatric Surgery, Oslo University Hospital, Rikshopitalet, Postboks 4950 Nydalen, 0424, Oslo, Norway.
| | | | - Åsmund Hermansen
- Department of Social Work, Child Welfare and Social Policy, Faculty of Social Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Marie Hamilton Larsen
- Department of Behavioral Medicine, Institute of Basic Medical Science, Faculty of Medicine, University of Oslo, Oslo, Norway
- Lovisenberg Diaconal University College, Oslo, Norway
| | - Kristin Bjørnland
- Department of Gastrointestinal and Pediatric Surgery, Oslo University Hospital, Rikshopitalet, Postboks 4950 Nydalen, 0424, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
15
|
Kleinau G, Chini B, Andersson L, Scheerer P. The role of G protein-coupled receptors and their ligands in animal domestication. Anim Genet 2024; 55:893-906. [PMID: 39324206 DOI: 10.1111/age.13476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
The domestication of plants and animals has resulted in one of the most significant cultural and socio-economical transitions in human history. Domestication of animals, including human-supervised reproduction, largely uncoupled particular animal species from their natural, evolutionary history driven by environmental and ecological factors. The primary motivations for domesticating animals were, and still are, producing food and materials (e.g. meat, eggs, honey or milk products, wool, leather products, jewelry and medication products) to support plowing in agriculture or in transportation (e.g. horse, cattle, camel and llama) and to facilitate human activities (for hunting, rescuing, therapeutic aid, guarding behavior and protecting or just as a companion). In recent years, decoded genetic information from more than 40 domesticated animal species have become available; these studies have identified genes and mutations associated with specific physiological and behavioral traits contributing to the complex genetic background of animal domestication. These breeding-altered genomes provide insights into the regulation of different physiological areas, including information on links between e.g. endocrinology and behavior, with important pathophysiological implications (e.g. for obesity and cancer), extending the interest in domestication well beyond the field. Several genes that have undergone selection during domestication and breeding encode specific G protein-coupled receptors, a class of membrane-spanning receptors involved in the regulation of a number of overarching functions such as reproduction, development, body homeostasis, metabolism, stress responses, cognition, learning and memory. Here we summarize the available literature on variations in G protein-coupled receptors and their ligands and how these have contributed to animal domestication.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Bice Chini
- CNR, Institute of Neuroscience, Vedano al Lambro, Italy, and NeuroMI - Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, USA
| | - Patrick Scheerer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| |
Collapse
|
16
|
Burns AJ, Goldstein AM. Causes and consequences: development and pathophysiology of Hirschsprung disease. WORLD JOURNAL OF PEDIATRIC SURGERY 2024; 7:e000903. [PMID: 39600627 PMCID: PMC11590806 DOI: 10.1136/wjps-2024-000903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Hirschsprung disease (HSCR) is a congenital enteric neuropathy in which the enteric nervous system (ENS) fails to develop along variable lengths of the distal gastrointestinal (GI) tract. This aganglionosis results in a functional bowel obstruction and requires surgical resection of the aganglionic segment. Despite surgery, however, long-term bowel dysfunction affects many patients. Understanding the embryologic causes and pathophysiologic consequences of HSCR is critical to improving its diagnosis and treatment. During normal gut development, the ENS arises from neural crest cells (NCCs) that delaminate from the neural tube to populate the entire GI tract with enteric neurons and glia. This process requires NCCs to undergo proliferation, migration and differentiation to form the complex neuroglial network that regulates gut motility and other intestinal functions. This review discusses the cellular and molecular processes that control normal ENS formation and what goes awry to give rise to HSCR. The complex pathophysiologic consequences of aganglionosis are discussed, including recent observations that describe novel aspects of HSCR beyond the absence of ganglion cells. This review aims to expand the understanding of HSCR and to stimulate new ideas on how to improve current management of the disease.
Collapse
Affiliation(s)
- Alan J Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Rogu PJ, Colalillo E, Rogu N, Rogu G. Atypical Presentation of Hirschsprung's Disease in an Infant: Challenges in Early Diagnosis and Management of Recurrent Enterocolitis. Cureus 2024; 16:e73862. [PMID: 39691132 PMCID: PMC11651778 DOI: 10.7759/cureus.73862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2024] [Indexed: 12/19/2024] Open
Abstract
Hirschsprung's disease is a congenital condition characterized by the absence of ganglion cells in the intestines, leading to bowel obstruction. The lack of ganglion cells disrupts the normal motility of the intestines, causing a functional obstruction. This can lead to enterocolitis, an inflammation of the intestines, which is a serious complication in infants with Hirschsprung's disease. This case report follows a male infant with multiple admissions for recurrent enterocolitis secondary to undiagnosed Hirschsprung's disease with unresolving diarrhea. This study underscores the significance of early diagnosis and management, even in cases of atypical presentation, emphasizing the role of antibiotics, bowel decompression, and surgical intervention. The findings highlight the complexity of managing Hirschsprung's disease and offer insights into tailored approaches for optimizing patient outcomes.
Collapse
Affiliation(s)
- Peter J Rogu
- Medicine, New York Institute of Technology (NYIT) College of Osteopathic Medicine, Old Westbury, USA
| | - Emily Colalillo
- Medicine, New York Institute of Technology (NYIT) College of Osteopathic Medicine, Old Westbury, USA
| | - Nicholas Rogu
- Pediatric Medicine, New York Institute of Technology (NYIT) College of Osteopathic Medicine, Old Westbury, USA
| | - George Rogu
- Pediatric Medicine, New York Institute of Technology (NYIT) College of Osteopathic Medicine, Old Westbury, USA
| |
Collapse
|
18
|
Zhong W, Lan C, Chen Y, Song K, Ma Z, Zeng J, Huang L, Zhang Y, Zhu Y, Xia H. Virus-Triggered Autoimmunity Was Associated With Hirschsprung's Disease Through Activation of Innate Immunity. J Immunol Res 2024; 2024:4838514. [PMID: 39493374 PMCID: PMC11531361 DOI: 10.1155/2024/4838514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 08/06/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Background: Hirschsprung's disease (HSCR) is a congenital enteric nervous system (ENS) disorder. Genetics cannot explain most sporadic cases. To explore the relationship between pathogen infection, autoantibodies, innate immune, and HSCR. Methods: Pathogen microarray was conducted in the serum of the prospective neonatal abdominal distension (NAD) cohort, consisting of 56 children followed for at least 6 months until the final diagnosis of HSCR was determined or excluded. We conducted an autoantibody microarray in an HSCR cohort, which is comprised of diagnosed HSCR patients (HSCR) and healthy control subjects (HC). RNA-seq of colon tissues from aganglionic and ganglionic segments of HSCR patients was performed. Results: Experimental results show that the serum lgM and lgG of enterovirus 71 (EV71) were significantly higher in HSCR than in the gastrointestinal dysfunction (GI) group, with a prediagnose value reaching area under the curve (AUC) over 0.76. We discovered that a group of autoantibodies were significantly higher in HSCR including neuronal pentraxin 1 (NPTX1), amyloid, neuron lysate, and myelin-associated oligodendrocytic basic protein (MOBP) than that in the HC group. These four autoantibodies could distinguish HSCR from the HC group, with a combined AUC of over 0.90 using both serum IgG and IgM. Further analysis showed that wide activation of innate immune pathways, including toll-like receptor (TLR) signaling pathway, neutrophil-to-lymphocyte ratio (NLR) signaling pathway, red cell distribution width to lymphocyte ratio (RLR) signaling pathway, and cyclic adenosine monophosphate (cAMP) signaling pathway in aganglionic compared to ganglionic segments of HSCR. Conclusion: This study suggested that virus-triggered autoimmunity may contribute to HSCR through activation of innate immunity, which facilitates the diagnosis and prevention of HSCR.
Collapse
Affiliation(s)
- Weiyong Zhong
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Chaoting Lan
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yuqiong Chen
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Kai Song
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Zuyi Ma
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Jixiao Zeng
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Lihua Huang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yan Zhang
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Yun Zhu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangdong Provincial Children's Medical Research Center, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| |
Collapse
|
19
|
Fujiwara N, Lee D, Li B, Pierro A, Yamataka A. Enhancement of enteric neural stem cell neurogenesis by glial cell-derived neurotrophic factor in experimental Hirschsprung's disease. Pediatr Surg Int 2024; 40:274. [PMID: 39460767 DOI: 10.1007/s00383-024-05861-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
PURPOSE Stem cell therapy offers a promising solution for congenital diseases like Hirschsprung's disease (HSCR). Optimizing stem cell efficacy by modifying the cells and their environment is crucial, but in vitro culture conditions need to be further improved. Glial cell-derived neurotrophic factor (GDNF) plays an important role in neuronal survival, proliferation, migration and differentiation during enteric nervous system (ENS) development. In this study, the effects of GDNF on neurites derived from an Ednrb knockout model were investigated with the aim of enhancing the neurogenic potential of enteric neural crest cells (ENCCs). METHODS Neurospheres were generated form Ednrb+/+ (control) and Ednrb-/- mice at embryonic day13.5 (E13.5) with Sox10-green fluorescent protein (Venus) transgenic expression. These neurospheres were cultured in control media and neurospheres from Ednrb-/- were cultured with either control media or media supplemented with GDNF. ENCCs differentiation was assessed using immunofluorescence staining after 18 days. RESULTS GDNF-treated Ednrb-/- neurospheres showed increased size and higher density of Sox10-positive ENCCs compared to untreated Ednrb-/- neurospheres. GDNF also enhanced the distribution of both TUJ1-positive neurons and S100-positive glial cells. CONCLUSION GDNF effectively enhanced the neurogenic potential of ENCCs from HSCR animal model. This finding is crucial for the development of cell therapy in HSCR.
Collapse
Affiliation(s)
- Naho Fujiwara
- Department of Pediatric General and Urogenital Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada.
| | - Dorothy Lee
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Canada
| | - Atsuyuki Yamataka
- Department of Pediatric General and Urogenital Surgery, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
20
|
Eshel Fuhrer A, Govorukhina O, Becker G, Ben-Shahar Y, Moran-Lev H, Sukhotnik I. Extended total colonic aganglionosis and total intestinal aganglionosis: Challenging enemies. Acta Paediatr 2024; 113:2297-2303. [PMID: 38949557 DOI: 10.1111/apa.17341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
AIM Extended total colonic aganglionosis (ETCA) represents uncommon forms of Hirschsprung disease (HD), with aganglionosis extending into the proximal small bowel. ETCA management is challenging and associated with poor outcomes and high mortality. This study compares management and outcomes of ETCA to more common HD forms. METHODS A retrospective cohort of HD patients (2012-2023) from two institutions. Three HD forms were compared: short-segment HD (SSHD, n = 19), long-segment HD or total colonic aganglionosis (LS/TCA, n = 9) and ETCA (n = 7). RESULTS Normally innervated segments in ETCA patients ranged 0-70 cm. Median times to first surgery were; ETCA = 3 days versus TCA = 21 days (p = 0.017) and SSHD = 95 days (p < 0.001), respectively. Median number of surgeries were; ETCA = 4, versus TCA = 2 (p = 0.17) and SSHD = 1 (p = 0.002), respectively. All the patients underwent a definitive pull-through procedure, except four ETCA patients with a permanent jejunostomy and residual aganglionic segment of 57-130 cm. ETCA patients had 92% lower odds of enterocolitis (14%) compared to TCA patients (67%, p = 0.054), and comparable odds to SSHD patients (16%, p = 0.92). ETCA mortality was 14%. CONCLUSION Extended total colonic aganglionosis patients require earlier and multiple interventions. Leaving an aganglionic segment may be advantageous, without increasing risk for enterocolitis. Tailored surgical treatment and rehabilitation programmes may prevent mortality and need for transplantation.
Collapse
Affiliation(s)
- Audelia Eshel Fuhrer
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Olga Govorukhina
- Republican Scientific and Practical Center for Pediatric Surgery, Minsk, Belarus
| | - Gal Becker
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Yoav Ben-Shahar
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Hadar Moran-Lev
- Pediatric Gastroenterology Unit, Dana-Dwek Children's Hospital, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Igor Sukhotnik
- Department of Pediatric Surgery, Dana-Dwek Children's Hospital, Affiliated to the Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| |
Collapse
|
21
|
Schermoly TP, Schropp KP. Colonic atresia and Hirschsprung's disease in a neonate: A case report. Int J Surg Case Rep 2024; 123:110250. [PMID: 39265370 PMCID: PMC11416630 DOI: 10.1016/j.ijscr.2024.110250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION Colonic atresia is a rare form of intestinal atresia that can be encountered in neonates. Although uncommon, other disease processes can be found simultaneously including malrotation, additional atresias, gastroschisis, and Hirschsprung's disease. CASE PRESENTATION A 2-day-old female neonate with known maternal polysubstance use was found to have colonic atresia on contrast enema after emesis and failure to pass meconium. Abdominal exploration revealed a blind ending cecum with evidence of ischemia along with an atretic transverse colon. An ileocecectomy with end ileostomy and transverse colon mucous fistula creation were performed. After eventual ileostomy reversal at 5 weeks of age, she struggled with intermittent oral intolerance and inconsistent bowel function. Re-exploration with ileostomy and gastrostomy tube placement was performed with additional biopsies revealing Hirschsprung's disease. CLINICAL DISCUSSION Concomitant colonic atresia and Hirschsprung's disease is a rare clinical entity that provides challenges in diagnosis and definitive surgical management. The suspected source of atresia in this case was presumed to be due to an intra-uterine vascular accident given maternal polysubstance use. Delays in diagnosis can lead to increased patient morbidity. CONCLUSION Even with a clear suspected etiology for colonic atresia, surgeons must maintain a high clinical suspicion for additional pathologies including but not limited to Hirschsprung's disease. Rectal suction biopsies should be performed if clinical suspicion arises for Hirschsprung's disease.
Collapse
Affiliation(s)
- Thomas P Schermoly
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66103, USA.
| | - Kurt P Schropp
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
22
|
Zhang Y, Zheng WH, Zhou SH, Gu JL, Yu Q, Zhu YZ, Yan YJ, Zhu Z, Shang JB. Molecular genetics, therapeutics and RET inhibitor resistance for medullary thyroid carcinoma and future perspectives. Cell Commun Signal 2024; 22:460. [PMID: 39342195 PMCID: PMC11439284 DOI: 10.1186/s12964-024-01837-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Medullary thyroid carcinoma (MTC) is a rare type of thyroid malignancy that accounts for approximately 1-2% of all thyroid cancers (TCs). MTC include hereditary and sporadic cases, the former derived from a germline mutation of rearrangement during transfection (RET) proto-oncogene, whereas somatic RET mutations are frequently present in the latter. Surgery is the standard treatment for early stage MTC, and the 10-year survival rate of early MTC is over 80%. While for metastatic MTC, chemotherapy showing low response rate, and there was a lack of effective systemic therapies in the past. Due to the high risk (ca. 15-20%) of distant metastasis and limited systemic therapies, the 10-year survival rate of patients with advanced MTC was only 10-40% from the time of first metastasis. Over the past decade, targeted therapy for RET has developed rapidly, bringing hopes to patients with advanced and progressive MTC. Two multi-kinase inhibitors (MKIs) including Cabozantinib and Vandetanib have been shown to increase progression-free survival (PFS) for patients with metastatic MTC and have been approved as choices of first-line treatment. However, these MKIs have not prolonged overall survival (OS) and their utility is limited due to high rates of off-target toxicities. Recently, new generation TKIs, including Selpercatinib and Pralsetinib, have demonstrated highly selective efficacy against RET and more favorable side effect profiles, and gained approval as second-line treatment options. Despite the ongoing development of RET inhibitors, the management of advanced and progressive MTC remains challenging, drug resistance remains the main reason for treatment failure, and the mechanisms are still unclear. Besides, new promising therapeutic approaches, such as novel drug combinations and next generation RET inhibitors are under development. Herein, we overview the pathogenesis, molecular genetics and current management approaches of MTC, and focus on the recent advances of RET inhibitors, summarize the current situation and unmet needs of these RET inhibitors in MTC, and provide an overview of novel strategies for optimizing therapeutic effects.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei-Hui Zheng
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shi-Hong Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jia-Lei Gu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang, China
| | - Qing Yu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yi-Zhou Zhu
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yu-Jie Yan
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Jin-Biao Shang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang, China.
| |
Collapse
|
23
|
Fries LE, Dharma S, Chakravarti A, Chatterjee S. Variability in proliferative and migratory defects in Hirschsprung disease-associated RET pathogenic variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614825. [PMID: 39372753 PMCID: PMC11451626 DOI: 10.1101/2024.09.24.614825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Despite the extensive genetic heterogeneity of Hirschsprung disease (HSCR; congenital colonic aganglionosis) 72% of patients harbor pathogenic variants in 10 genes that form a gene regulatory network (GRN) controlling the development of the enteric nervous system (ENS). Among these genes, the receptor tyrosine kinase gene RET is the most significant contributor, accounting for pathogenic variants in 12%-50% of patients depending on phenotype. RET plays a critical role in the proliferation and migration of ENS precursors, and defects in these processes lead to HSCR. However, despite the gene's importance in HSCR, the functional consequences of RET pathogenic variants and their mechanism of disease remain poorly understood. To address this, we investigated the proliferative and migratory phenotypes in a RET-dependent neural crest-derived cell line harboring one of five missense (L56M, E178Q, Y791F, S922Y, F998L) or three nonsense (Y204X, R770X, Y981X) pathogenic heterozygous variants. Using a combination of cDNA-based and CRISPR-based PRIME editing coupled with quantitative proliferation and migration assays, we detected significant losses in cell proliferation and migration in three missense (E178Q, S922Y, F998L) and all nonsense variants. Our data suggests that the Y791F variant, whose pathogenicity has been debated, is likely not pathogenic. Importantly, the severity of migration loss did not consistently correlate with proliferation defects, and the phenotypic severity of nonsense variants was independent of their position within the RET protein. This study highlights the necessity and feasibility of targeted functional assays to accurately assess the pathogenicity of HSCR-associated variants, rather than relying solely on machine learning predictions, which could themselves be refined by incorporating such functional data.
Collapse
Affiliation(s)
- Lauren E Fries
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016
| | - Sree Dharma
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016
| | - Aravinda Chakravarti
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Sumantra Chatterjee
- Center for Human Genetics & Genomics, New York University Grossman School of Medicine, New York, NY 10016
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
24
|
Dorkins H. JMG at 60. J Med Genet 2024; 61:823. [PMID: 39181713 DOI: 10.1136/jmg-2024-110287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Affiliation(s)
- Huw Dorkins
- St Peter's College, University of Oxford, Oxford, UK
- Editor in Chief, Journal of Medical Genetics
| |
Collapse
|
25
|
Jevans B, Cooper F, Fatieieva Y, Gogolou A, Kang YN, Restuadi R, Moulding D, Vanden Berghe P, Adameyko I, Thapar N, Andrews PW, De Coppi P, Tsakiridis A, McCann CJ. Human enteric nervous system progenitor transplantation improves functional responses in Hirschsprung disease patient-derived tissue. Gut 2024; 73:1441-1453. [PMID: 38816188 PMCID: PMC11347211 DOI: 10.1136/gutjnl-2023-331532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE Hirschsprung disease (HSCR) is a severe congenital disorder affecting 1:5000 live births. HSCR results from the failure of enteric nervous system (ENS) progenitors to fully colonise the gastrointestinal tract during embryonic development. This leads to aganglionosis in the distal bowel, resulting in disrupted motor activity and impaired peristalsis. Currently, the only viable treatment option is surgical resection of the aganglionic bowel. However, patients frequently suffer debilitating, lifelong symptoms, with multiple surgical procedures often necessary. Hence, alternative treatment options are crucial. An attractive strategy involves the transplantation of ENS progenitors generated from human pluripotent stem cells (hPSCs). DESIGN ENS progenitors were generated from hPSCs using an accelerated protocol and characterised, in detail, through a combination of single-cell RNA sequencing, protein expression analysis and calcium imaging. We tested ENS progenitors' capacity to integrate and affect functional responses in HSCR colon, after ex vivo transplantation to organotypically cultured patient-derived colonic tissue, using organ bath contractility. RESULTS We found that our protocol consistently gives rise to high yields of a cell population exhibiting transcriptional and functional hallmarks of early ENS progenitors. Following transplantation, hPSC-derived ENS progenitors integrate, migrate and form neurons/glia within explanted human HSCR colon samples. Importantly, the transplanted HSCR tissue displayed significantly increased basal contractile activity and increased responses to electrical stimulation compared with control tissue. CONCLUSION Our findings demonstrate, for the first time, the potential of hPSC-derived ENS progenitors to repopulate and increase functional responses in human HSCR patient colonic tissue.
Collapse
Affiliation(s)
- Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL GOS Institute of Child Health, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Fay Cooper
- School of Biosciences, The University of Sheffield, Sheffield, UK
- Neuroscience Institute, The University of Sheffield, Sheffield, UK
| | - Yuliia Fatieieva
- Department of Neuroimmunology, Centre for Brain Research, Medical University of Vienna, Wien, Austria
| | - Antigoni Gogolou
- School of Biosciences, The University of Sheffield, Sheffield, UK
- Neuroscience Institute, The University of Sheffield, Sheffield, UK
| | - Yi-Ning Kang
- Laboratory for Enteric NeuroScience (LENS), Translational Research Centre for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Restuadi Restuadi
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Dale Moulding
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Centre for Gastrointestinal Disorders (TARGID), Katholieke Universiteit Leuven, Leuven, Belgium
- Cell and Tissue Imaging Cluster (CIC), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Igor Adameyko
- Department of Neuroimmunology, Centre for Brain Research, Medical University of Vienna, Wien, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL GOS Institute of Child Health, London, UK
- Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital UQ Faculty, South Brisbane, Queensland, Australia
| | - Peter W Andrews
- School of Biosciences, The University of Sheffield, Sheffield, UK
- Neuroscience Institute, The University of Sheffield, Sheffield, UK
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine, UCL GOS Institute of Child Health, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Specialist Neonatal and Paediatric Surgery Unit, Great Ormond Street Hospital, London, UK
| | - Anestis Tsakiridis
- School of Biosciences, The University of Sheffield, Sheffield, UK
- Neuroscience Institute, The University of Sheffield, Sheffield, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL GOS Institute of Child Health, London, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
26
|
Maselli KM, Shah NR, Speck KE. Approach to Constipation in Children: Recommendations for Evaluation and Management. Adv Pediatr 2024; 71:195-211. [PMID: 38944484 DOI: 10.1016/j.yapd.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Constipation is common in childhood, and most patients can be successfully managed by their primary care provider. However, some patients will require more specialized management either due to an underlying congenital colorectal disorder such as Hirschsprung disease or anorectal malformation or due to severe functional constipation that is refractory to medical management.
Collapse
Affiliation(s)
- Kathryn M Maselli
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, C.S. Mott Children's Hospital, 1540 E. Hospital Drive, Ann Arbor, MI 48109-4211, USA
| | - Nikhil R Shah
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, C.S. Mott Children's Hospital, 1540 E. Hospital Drive, Ann Arbor, MI 48109-4211, USA
| | - Karen Elizabeth Speck
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, C.S. Mott Children's Hospital, 1540 E. Hospital Drive, Ann Arbor, MI 48109-4211, USA.
| |
Collapse
|
27
|
Karkala F, Wang M, Alves MM, Parikh K. Intestinal Motility May Not Be Completely Dependent on Epithelial RET Signaling in the Adult Intestine. Gastroenterology 2024; 167:629-630. [PMID: 38593923 DOI: 10.1053/j.gastro.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Affiliation(s)
- Faidra Karkala
- Departments of Clinical Genetics and Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Min Wang
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Maria M Alves
- Departments of Clinical Genetics and Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Kaushal Parikh
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Mombeek LMM, Boesmans W, Wilson DM. Genomic stress in diseases stemming from defects in the second brain. Neurogastroenterol Motil 2024:e14860. [PMID: 39004995 DOI: 10.1111/nmo.14860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/04/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024]
Abstract
This review discusses the less-explored realm of DNA damage and repair within the enteric nervous system (ENS), often referred to as the "second brain." While the central nervous system has been extensively studied for its DNA repair mechanisms and associated neuropathologies, the ENS, which can autonomously coordinate gastrointestinal function, experiences unique challenges and vulnerabilities related to its genome integrity. The susceptibility of the ENS to DNA damage is exacerbated by its limited protective barriers, resulting in not only endogenous genotoxic exposures, such as oxidative stress, but also exogenous threats, such as ingested environmental contaminants, local inflammatory responses, and gut dysbiosis. Here, we discuss the evidence for DNA repair defects in enteric neuropathies, most notably, the reported relationship between inherited mutations in RAD21 and LIG3 with chronic intestinal pseudo-obstruction and mitochondrial gastrointestinal encephalomyopathy disorders, respectively. We also introduce the lesser-recognized gastrointestinal complications in DNA repair syndromes, including conditions like Cockayne syndrome. The review concludes by pointing out the potential role of DNA repair defects in not only congenital disorders but also aging-related gut dysfunction, as well as the crucial need for further research to establish direct causal links between DNA damage accumulation and ENS-specific pathologic phenotypes.
Collapse
Affiliation(s)
- Lobke Marie M Mombeek
- Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Werend Boesmans
- Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - David M Wilson
- Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
29
|
Morera C, Nurko S. Hirschsprung's disease. Management. Aliment Pharmacol Ther 2024; 60 Suppl 1:S66-S76. [PMID: 38922939 DOI: 10.1111/apt.18068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/08/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Hirschsprung's disease (HD) is a rare congenital disease that is characterised by the absence of ganglion cells in the myenteric plexus starting in the distal bowel. This results in distal functional obstruction and may lead to complications like enterocolitis. The treatment is surgical and requires the resection of the aganglionic segment, and the pull-through of normal intestine into the anal opening. However, even after successful surgery, patients may continue to have symptoms. AIM Discuss current surgical techniques and management strategies for patients with postoperative symptoms after surgical correction of Hirschsprung's disease. METHODS A review of the literature was done through PubMed, with a focus on clinical management and approach. RESULTS We describe the clinical problems that can occur after surgical correction. These include obstructive symptoms, enterocolitis, or faecal incontinence. A systematic approach for the evaluation of these patients includes the exclusion of anatomic, inflammatory, behavioural or motility related factors. Depending on the severity of the symptoms, the evaluation includes examination under anaesthesia, the performance of contrast studies, endoscopic studies, measurement of anal sphincter function and colonic motility studies. The treatment is focused towards addressing the different pathophysiological mechanisms, and may include medical management, botulinum toxin to the anal sphincter or rarely redo-operation. CONCLUSIONS Patients with Hirschsprung's disease need to have surgical correction, and their postoperative long-term management is complex given a variety of associated problems that can occur after surgery. A systematic evaluation is necessary to provide appropriate therapy.
Collapse
Affiliation(s)
- Claudio Morera
- Center for Motility and Functional Gastrointestinal Disorders, Children's Hospital Boston, Boston, Massachusetts, USA
| | - Samuel Nurko
- Center for Motility and Functional Gastrointestinal Disorders, Children's Hospital Boston, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Mariana N, Hatta M, Lia E, Lampus H, Prihantono, Russeng SS, Makkadafi M, Fadilat M, Husni MA, Faruk M. Toll-like receptor-8 mRNA expression as a predictor of enterocolitis in hirschsprung disease: a cross-sectional study. INTERNATIONAL JOURNAL OF SURGERY OPEN 2024; 62:401-405. [DOI: 10.1097/io9.0000000000000148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/10/2024] [Indexed: 01/12/2025]
Abstract
Introduction:
Hirschsprung-associated enterocolitis (HAEC) is an inflammatory intestinal condition occurring in patients with Hirschsprung disease (HD). While the exact pathogenesis remains unclear, factors such as intestinal mucosal barrier injury, abnormal immune responses, and pathogenic infections contribute to HAEC. Toll-like receptors (TLRs) 7 and 8 are key in viral recognition and correlate with intestinal mucosal inflammation. In particular, TLR-8 contributes to antiviral responses, and chronic inflammation, and can be targeted with adjuvants. This study investigates the relationship between TLR-8 mRNA expression and enterocolitis in HD.
Methods:
This cross-sectional study utilized simple random sampling. Histopathological analysis of colon samples from HD patients was used to assess enterocolitis severity, and RT-PCR quantified TLR-8 mRNA expression. HD diagnosis was confirmed via rectal biopsy, and enterocolitis was graded using Teitelbaum’s classification.
Results:
The study included 29 patients with an average age of 4.8 years. TLR-8 mRNA expression averaged 8.5 (fold change), ranging from 6.5 to 13.8. Subjects were divided into low (≤8.1) and high (>8.1) expression groups, with 15 (51.7%) and 14 (48.3%) patients, respectively. Enterocolitis severity distribution was 22 (75.9%) normal, 1 (3.4%) grade II, 4 (13.8%) grade III, and 2 (6.9%) IV.
Conclusion:
There is a significant association between TLR-8 mRNA expression and the incidence of enterocolitis in patients with HD. The authors found that increased TLR-8 mRNA expression in HD patients resulted in a twofold higher risk of enterocolitis. Altogether, our data indicates the potential for TLR-8 mRNA expression in biopsies as an indicator of the risk of enterocolitis development in patients with HD.
Collapse
Affiliation(s)
- Nita Mariana
- Department of Pediatric Surgery, Faculty of Medicine, Hasanuddin University – Dr. Wahidin Sudirohusodo Hospital, Makassar
| | - Mochammad Hatta
- Department of Clinical Microbiology, Faculty of Medicine, Hasanuddin University – Hasanuddin University Hospital, Makassar
| | - Emiliana Lia
- Department of Pediatric Surgery, Faculty of Medicine, Padjadjaran University, Bandung
| | - Harsali Lampus
- Department of Surgery, Faculty of Medicine, Sam Ratulangi University, Manado
| | - Prihantono
- Department of Surgery, Faculty of Medicine, Hasanuddin University – Hasanuddin University Hospital, Makassar
| | - Syamsiar S. Russeng
- Department of Occupational Health and Safety, Faculty of Public Health, Hasanuddin University, Makassar
| | - Munawir Makkadafi
- Department of Pediatric Surgery, Faculty of Medicine, Hasanuddin University – Hasanuddin University Hospital, Makassar
| | - Marwah Fadilat
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Muh. Amrul Husni
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Muhammad Faruk
- Department of Surgery, Faculty of Medicine, Hasanuddin University – Hasanuddin University Hospital, Makassar
| |
Collapse
|
31
|
Malbos M, Wakeling E, Gautier T, Boespflug-Tanguy O, Busby L, Taylor-Miller T, Dudoignon B, Bokov P, Govin J, Grisval M, Rega A, Mourot De Rougemont MG, Aubriot-Lorton MH, Darmency V, Bensignor C, Houzel A, Huet F, Denommé-Pichon AS, Delanne J, Tran Mau-Them F, Bruel AL, Safraou H, Nambot S, Garde A, Philippe C, Duffourd Y, Vitobello A, Faivre L, Thauvin-Robinet C. Further description of two individuals with de novo p.(Glu127Lys) missense variant in the ASCL1 gene. Clin Genet 2024; 105:555-560. [PMID: 38287449 DOI: 10.1111/cge.14485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/31/2024]
Abstract
Achaete-Scute Family basic-helix-loop-helix (bHLH) Transcription Factor 1 (ASCL1) is a proneural transcription factor involved in neuron development in the central and peripheral nervous system. While initially suspected to contribute to congenital central hypoventilation syndrome-1 (CCHS) with or without Hirschsprung disease (HSCR) in three individuals, its implication was ruled out by the presence, in one of the individuals, of a Paired-like homeobox 2B (PHOX2B) heterozygous polyalanine expansion variant, known to cause CCHS. We report two additional unrelated individuals sharing the same sporadic ASCL1 p.(Glu127Lys) missense variant in the bHLH domain and a common phenotype with short-segment HSCR, signs of dysautonomia, and developmental delay. One has also mild CCHS without polyalanine expansion in PHOX2B, compatible with the diagnosis of Haddad syndrome. Furthermore, missense variants with homologous position in the same bHLH domain in other genes are known to cause human diseases. The description of additional individuals carrying the same variant and similar phenotype, as well as targeted functional studies, would be interesting to further evaluate the role of ASCL1 in neurocristopathies.
Collapse
Affiliation(s)
- Marlène Malbos
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
| | - Emma Wakeling
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Thierry Gautier
- Université Grenoble Alpes, Inserm-U1209, CNRS-UMR5309, Institut pour l'Avancée des Biosciences, Grenoble, France
| | - Odile Boespflug-Tanguy
- Université Paris-Cité, INSERM-UMR1141, CRMR « Leucodystrophies », Neurologie Pédiatrique et Maladies métaboliques, Hôpital Robert-Debré, AP-HP, Paris, France
| | - Louise Busby
- Rare & Inherited Disease Laboratory, London North Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Tashunka Taylor-Miller
- Rare & Inherited Disease Laboratory, London North Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Benjamin Dudoignon
- Université Paris-Cité, AP-HP, Hôpital Robert-Debré, Physiologie Pédiatrique-Centre du Sommeil-CRMR Hypoventilations alvéolaires rares, INSERM, Paris, France
| | - Plamen Bokov
- Université Paris-Cité, AP-HP, Hôpital Robert-Debré, Physiologie Pédiatrique-Centre du Sommeil-CRMR Hypoventilations alvéolaires rares, INSERM, Paris, France
| | - Jérôme Govin
- Université Grenoble Alpes, Inserm-U1209, CNRS-UMR5309, Institut pour l'Avancée des Biosciences, Grenoble, France
| | - Margot Grisval
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
| | | | | | | | | | - Candace Bensignor
- CCMR "Maladies Endocriniennes de la Croissance et du Développement", CHU Dijon, Dijon, France
| | - Anne Houzel
- Pneumologie Pédiatrique, CHU Dijon, Dijon, France
| | - Frédéric Huet
- Pédiatrie pluridisciplinaire, CHU Dijon, Dijon, France
| | - Anne-Sophie Denommé-Pichon
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
- Inserm-UB-UMR1231 GAD, Dijon, France
| | - Julian Delanne
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
| | - Frédéric Tran Mau-Them
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
- Inserm-UB-UMR1231 GAD, Dijon, France
| | - Ange-Line Bruel
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
- Inserm-UB-UMR1231 GAD, Dijon, France
| | - Hana Safraou
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
- Inserm-UB-UMR1231 GAD, Dijon, France
| | - Sophie Nambot
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
| | - Aurore Garde
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
| | - Christophe Philippe
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
- Inserm-UB-UMR1231 GAD, Dijon, France
| | | | - Antonio Vitobello
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
- Inserm-UB-UMR1231 GAD, Dijon, France
| | - Laurence Faivre
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
| | - Christel Thauvin-Robinet
- CRMRs "Anomalies du Développement et syndromes malformatifs" et "Déficiences Intellectuelles de causes rares", Centre de Génétique, CHU Dijon, Dijon, France
- UF "Innovation diagnostique dans les maladies rares", CHU Dijon, Dijon, France
| |
Collapse
|
32
|
Zhang Y, Song F, Yang M, Chen C, Cui J, Xing M, Dai Y, Li M, Cao Y, Lu L, Zhu H, Liu Y, Ma C, Wei Q, Qin H, Li J. Gastrointestinal Dysmotility Predisposes to Colitis through Regulation of Gut Microbial Composition and Linoleic Acid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306297. [PMID: 38477534 PMCID: PMC11132037 DOI: 10.1002/advs.202306297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Indexed: 03/14/2024]
Abstract
Disrupted gastrointestinal (GI) motility is highly prevalent in patients with inflammatory bowel disease (IBD), but its potential causative role remains unknown. Herein, the role and the mechanism of impaired GI motility in colitis pathogenesis are investigated. Increased colonic mucosal inflammation is found in patients with chronic constipation (CC). Mice with GI dysmotility induced by genetic mutation or chemical insult exhibit increased susceptibility to colitis, dependent on the gut microbiota. GI dysmotility markedly decreases the abundance of Lactobacillus animlalis and increases the abundance of Akkermansia muciniphila. The reduction in L. animlalis, leads to the accumulation of linoleic acid due to compromised conversion to conjugated linoleic acid. The accumulation of linoleic acid inhibits Treg cell differentiation and increases colitis susceptibility via inducing macrophage infiltration and proinflammatory cytokine expression in macrophage. Lactobacillus and A. muciniphila abnormalities are also observed in CC and IBD patients, and mice receiving fecal microbiota from CC patients displayed an increased susceptibility to colitis. These findings suggest that GI dysmotility predisposes host to colitis development by modulating the composition of microbiota and facilitating linoleic acid accumulation. Targeted modulation of microbiota and linoleic acid metabolism may be promising to protect patients with motility disorder from intestinal inflammation.
Collapse
Affiliation(s)
- Youhua Zhang
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Feifei Song
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Muqing Yang
- Department of General SurgeryShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Chunqiu Chen
- Diagnostic and Treatment Center for Refractory Diseases of Abdomen SurgeryShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Jiaqu Cui
- Department of Colorectal DiseaseShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Mengyu Xing
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Yuna Dai
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Man Li
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Yuan Cao
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Ling Lu
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Huiyuan Zhu
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Ying Liu
- Department of General SurgeryShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Chunlian Ma
- Department of Colorectal DiseaseShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Qing Wei
- Department of PathologyShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Huanlong Qin
- Department of Gastrointestinal SurgeryShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
| | - Jiyu Li
- Department of General SurgeryShanghai Tenth People's Hospital, Tongji University School of MedicineShanghai200072China
- Geriatric Cancer CenterHuaDong Hospital Affiliated to Fudan
UniversityShanghai200040China
| |
Collapse
|
33
|
Wang ZX, Li QQ, Cai J, Wu JZ, Wang JJ, Zhang MY, Wang QX, Tong ZJ, Yang J, Wei TH, Zhou Y, Dai WC, Ding N, Leng XJ, Sun SL, Xue X, Yu YC, Yang Y, Li NG, Shi ZH. Unraveling the Promise of RET Inhibitors in Precision Cancer Therapy by Targeting RET Mutations. J Med Chem 2024; 67:4346-4375. [PMID: 38484122 DOI: 10.1021/acs.jmedchem.3c02319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Over the past decades, the role of rearranged during transfection (RET) alterations in tumorigenesis has been firmly established. RET kinase inhibition is an essential therapeutic target in patients with RET-altered cancers. In clinical practice, initial efficacy can be achieved in patients through the utilization of multikinase inhibitors (MKIs) with RET inhibitory activity. However, the effectiveness of these MKIs is impeded by the adverse events associated with off-target effects. Recently, many RET-selective inhibitors, characterized by heightened specificity and potency, have been developed, representing a substantial breakthrough in the field of RET precision oncology. This Perspective focuses on the contemporary understanding of RET mutations, recent advancements in next-generation RET inhibitors, and the challenges associated with resistance to RET inhibitors. It provides valuable insights for the development of next-generation MKIs and selective RET inhibitors.
Collapse
Affiliation(s)
- Zi-Xuan Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Qing-Qing Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jiao Cai
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jia-Zhen Wu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jing-Jing Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Meng-Yuan Zhang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Qing-Xin Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhen-Jiang Tong
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Jin Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yun Zhou
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Wei-Chen Dai
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Ning Ding
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Xue-Jiao Leng
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Xin Xue
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yan-Cheng Yu
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| |
Collapse
|
34
|
Dershowitz LB, Kaltschmidt JA. Enteric Nervous System Striped Patterning and Disease: Unexplored Pathophysiology. Cell Mol Gastroenterol Hepatol 2024; 18:101332. [PMID: 38479486 PMCID: PMC11176954 DOI: 10.1016/j.jcmgh.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
The enteric nervous system (ENS) controls gastrointestinal (GI) motility, and defects in ENS development underlie pediatric GI motility disorders. In disorders such as Hirschsprung's disease (HSCR), pediatric intestinal pseudo-obstruction (PIPO), and intestinal neuronal dysplasia type B (INDB), ENS structure is altered with noted decreased neuronal density in HSCR and reports of increased neuronal density in PIPO and INDB. The developmental origin of these structural deficits is not fully understood. Here, we review the current understanding of ENS development and pediatric GI motility disorders incorporating new data on ENS structure. In particular, emerging evidence demonstrates that enteric neurons are patterned into circumferential stripes along the longitudinal axis of the intestine during mouse and human development. This novel understanding of ENS structure proposes new questions about the pathophysiology of pediatric GI motility disorders. If the ENS is organized into stripes, could the observed changes in enteric neuron density in HSCR, PIPO, and INDB represent differences in the distribution of enteric neuronal stripes? We review mechanisms of striped patterning from other biological systems and propose how defects in striped ENS patterning could explain structural deficits observed in pediatric GI motility disorders.
Collapse
Affiliation(s)
- Lori B Dershowitz
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California; Wu Tsai Neurosciences Institute, Stanford University, Stanford, California
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California; Wu Tsai Neurosciences Institute, Stanford University, Stanford, California.
| |
Collapse
|
35
|
Kuil LE, Chauhan RK, de Graaf BM, Cheng WW, Kakiailatu NJM, Lasabuda R, Verhaeghe C, Windster JD, Schriemer D, Azmani Z, Brooks AS, Edie S, Reeves RH, Eggen BJL, Shepherd IT, Burns AJ, Hofstra RMW, Melotte V, Brosens E, Alves MM. ATP5PO levels regulate enteric nervous system development in zebrafish, linking Hirschsprung disease to Down Syndrome. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166991. [PMID: 38128843 DOI: 10.1016/j.bbadis.2023.166991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Hirschsprung disease (HSCR) is a complex genetic disorder characterized by the absence of enteric nervous system (ENS) in the distal region of the intestine. Down Syndrome (DS) patients have a >50-fold higher risk of developing HSCR than the general population, suggesting that overexpression of human chromosome 21 (Hsa21) genes contribute to HSCR etiology. However, identification of responsible genes remains challenging. Here, we describe a genetic screening of potential candidate genes located on Hsa21, using the zebrafish. Candidate genes were located in the DS-HSCR susceptibility region, expressed in the human intestine, were known potential biomarkers for DS prenatal diagnosis, and were present in the zebrafish genome. With this approach, four genes were selected: RCAN1, ITSN1, ATP5PO and SUMO3. However, only overexpression of ATP5PO, coding for a component of the mitochondrial ATPase, led to significant reduction of ENS cells. Paradoxically, in vitro studies showed that overexpression of ATP5PO led to a reduction of ATP5PO protein levels. Impaired neuronal differentiation and reduced mitochondrial ATP production, were also detected in vitro, after overexpression of ATP5PO in a neuroblastoma cell line. Finally, epistasis was observed between ATP5PO and ret, the most important HSCR gene. Taken together, our results identify ATP5PO as the gene responsible for the increased risk of HSCR in DS patients in particular if RET variants are also present, and show that a balanced expression of ATP5PO is required for normal ENS development.
Collapse
Affiliation(s)
- L E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - R K Chauhan
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - B M de Graaf
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - W W Cheng
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - N J M Kakiailatu
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - R Lasabuda
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - C Verhaeghe
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - J D Windster
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center Rotterdam, Sophia's Children's Hospital, Rotterdam, the Netherlands
| | - D Schriemer
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Z Azmani
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - A S Brooks
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - S Edie
- Johns Hopkins University School of Medicine, Department of Physiology and McKusick-Nathans Department of Genetic Medicine, Baltimore, MD, United States of America
| | - R H Reeves
- Johns Hopkins University School of Medicine, Department of Physiology and McKusick-Nathans Department of Genetic Medicine, Baltimore, MD, United States of America
| | - B J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - I T Shepherd
- Department of Biology, Emory University, Atlanta, GA, United States of America
| | - A J Burns
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Birth Defects Research Centre, UCL Institute of Child Health, London, United Kingdom; Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals, Cambridge, MA, United States of America
| | - R M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - V Melotte
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pathology, GROW-school for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - E Brosens
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - M M Alves
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center Rotterdam, Sophia's Children's Hospital, Rotterdam, the Netherlands.
| |
Collapse
|
36
|
Uribe RA. Genetic regulation of enteric nervous system development in zebrafish. Biochem Soc Trans 2024; 52:177-190. [PMID: 38174765 PMCID: PMC10903509 DOI: 10.1042/bst20230343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
The enteric nervous system (ENS) is a complex series of interconnected neurons and glia that reside within and along the entire length of the gastrointestinal tract. ENS functions are vital to gut homeostasis and digestion, including local control of peristalsis, water balance, and intestinal cell barrier function. How the ENS develops during embryological development is a topic of great concern, as defects in ENS development can result in various diseases, the most common being Hirschsprung disease, in which variable regions of the infant gut lack ENS, with the distal colon most affected. Deciphering how the ENS forms from its progenitor cells, enteric neural crest cells, is an active area of research across various animal models. The vertebrate animal model, zebrafish, has been increasingly leveraged to understand early ENS formation, and over the past 20 years has contributed to our knowledge of the genetic regulation that underlies enteric development. In this review, I summarize our knowledge regarding the genetic regulation of zebrafish enteric neuronal development, and based on the most current literature, present a gene regulatory network inferred to underlie its construction. I also provide perspectives on areas for future zebrafish ENS research.
Collapse
Affiliation(s)
- Rosa A. Uribe
- Biosciences Department, Rice University, Houston, TX 77005, U.S.A
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, TX 77005, U.S.A
| |
Collapse
|
37
|
Sowulewski O, Bubińska M, Zagierska A, Zagierski M, Szlagatys-Sidorkiewicz A. High-Resolution Anorectal Manometry as a Screening Tool for Hirschsprung's Disease: A Comprehensive Retrospective Analysis. J Clin Med 2024; 13:1268. [PMID: 38592083 PMCID: PMC10932072 DOI: 10.3390/jcm13051268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/26/2024] [Accepted: 02/14/2024] [Indexed: 04/10/2024] Open
Abstract
Hirschsprung's disease (HD) is characterized by a congenital absence of enteric ganglion cells in the intestine, posing challenges in diagnosis, particularly in pediatric patients. The gold standard, rectal suction biopsy (RSB), carries risks, prompting an exploration of non-invasive alternatives such as high-resolution anorectal manometry (HR-ARM) for HD screening. We conducted a retrospective analysis of 136 patients suspected of HD between 2018 and 2022, which were stratified into three age groups: ≤12 months, ≤24 months, and >24 months. Criteria for suspicion included delayed meconium passage, unresponsive chronic constipation, and abnormal prior test results. HR-ARM, supplemented by additional tests, confirmed 16 HD cases. HR-ARM exhibited 93.75% sensitivity, 89.47% specificity, 99.03% negative predictive value (NPV), and 55.56% positive predictive value (PPV). Notably, HR-ARM consistently performed well in patients ≤ 2 years old but demonstrated reduced efficacy in older children, which was likely due to complications from chronic constipation. This study underscores HR-ARM's promise as a non-invasive HD screening tool, especially in younger patients. However, its limitations in older children warrant consideration. Establishing standardized protocols, particularly for assessing the recto-anal inhibitory reflex, is crucial. Further research is imperative to optimize HR-ARM's diagnostic role across varied age groups in HD assessment.
Collapse
Affiliation(s)
- Oliver Sowulewski
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.B.); (A.Z.); (M.Z.); (A.S.-S.)
| | | | | | | | | |
Collapse
|
38
|
Sheikh SS, Sharath HV, Seth NH. A Rare Case Report on Postoperative Rehabilitation in Hirschsprung Disease. Cureus 2024; 16:e54044. [PMID: 38481896 PMCID: PMC10933885 DOI: 10.7759/cureus.54044] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/12/2024] [Indexed: 03/03/2025] Open
Abstract
Hirschsprung disease (HD) is characterized by the absence of ganglion cells in the myenteric and submucosal plexuses of the hindgut. Here, we report a case of an eight-year-old male child who had abdominal distension with a history of repetitive gas passage and a complaint of stool passage. In February 2023, the patient was diagnosed with Hirshsprung disease, for which a left-side colostomy was done. In November 2023, he underwent Hirshsprung stage 2 repair. He was operated on the 17th of December 2023 under general anaesthesia colostomy mobilization. Physiotherapy commencement and evaluation were started on the 18th of December 2023. After the colostomy procedure, the incision weakened the abdominal and lower limb muscles, while bowel obstruction and discomfort further impeded the patient's ability to perform daily activities. Physical examination revealed increased work of breathing, reduced range of motion of the bilateral hip joint, reduced muscle strength of lower limb musculature, reduced abdominal muscle strength, difficulty in walking and waddling type of gait. Physiotherapy goals were set based on the problem list. The patient showed improvement in the two weeks of physiotherapy commencement, followed by improvement in functional ability. The comprehensive care provided during the rehabilitation phase aimed at addressing the specific needs arising from the surgical intervention, promoting optimal bowel function, improving ranges and strength and ensuring overall well-being.
Collapse
Affiliation(s)
- Shifa S Sheikh
- Department of Paediatric Physiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education & Research (Deemed to be University), Wardha, IND
| | - H V Sharath
- Department of Paediatric Physiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education & Research (Deemed to be University), Wardha, IND
| | - Nikita H Seth
- Department of Neurophysiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education & Research (Deemed to be University), Wardha, IND
| |
Collapse
|
39
|
Hergenreder T, Yang T, Ye B. The role of Down syndrome cell adhesion molecule in Down syndrome. MEDICAL REVIEW (2021) 2024; 4:31-41. [PMID: 38515781 PMCID: PMC10954295 DOI: 10.1515/mr-2023-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/18/2024] [Indexed: 03/23/2024]
Abstract
Down syndrome (DS) is caused by the presence of an extra copy of the entire or a portion of human chromosome 21 (HSA21). This genomic alteration leads to elevated expression of numerous HSA21 genes, resulting in a variety of health issues in individuals with DS. Among the genes located in the DS "critical region" of HSA21, Down syndrome cell adhesion molecule (DSCAM) plays an important role in neuronal development. There is a growing body of evidence underscoring DSCAM's involvement in various DS-related disorders. This review aims to provide a concise overview of the established functions of DSCAM, with a particular focus on its implications in DS. We delve into the roles that DSCAM plays in DS-associated diseases. In the concluding section of this review, we explore prospective avenues for future research to further unravel DSCAM's role in DS and opportunities for therapeutic treatments.
Collapse
Affiliation(s)
- Ty Hergenreder
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Tao Yang
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Bing Ye
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
40
|
Ding HY, Lei W, Xiao SJ, Deng H, Yuan LK, Xu L, Zhou JL, Huang R, Fang YL, Wang QY, Zhang Y, Zhang L, Zhu XC. High incidence of EDNRB gene mutation in seven southern Chinese familial cases with Hirschsprung's disease. Pediatr Surg Int 2024; 40:38. [PMID: 38253735 DOI: 10.1007/s00383-023-05620-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 01/24/2024]
Abstract
PURPOSE Hirschsprung's disease (HSCR) is the leading cause of neonatal functional intestinal obstruction, which has been identified in many familial cases. HSCR, a multifactorial disorder of enteric nervous system (ENS) development, is associated with at least 24 genes and seven chromosomal loci, with RET and EDNRB as its major genes. We present a genetic investigation of familial HSCR to clarify the genotype-phenotype relationship. METHODS We performed whole exome sequencing (WES) on Illumina HiSeq X Ten platform to investigate genetic backgrounds of core family members, and identified the possibly harmful mutation genes. Mutation carriers and pedigree relatives were validated by Sanger sequencing for evaluating the gene penetrance. RESULTS Four familial cases showed potential disease-relative variants in EDNRB and RET gene, accounting for all detection rate of 57.1%. Three familial cases exhibited strong pathogenic variants as frameshift or missense mutations in EDNRB gene. A novel c.367delinsTT mutation of EDNRB was identified in one family member. The other two EDNRB mutations, c.553G>A in family 2 and c.877delinsTT in family 5, have been reported in previous literatures. The penetrance of EDNRB variants was 33-50% according mutation carries. In family 6, the RET c.1858T>C (C620R) point mutation has previously been reported to cause HSCR, with 28.5% penetrance. CONCLUSION We identified a novel EDNRB (deleted C and inserted TT) mutation in this study using WES. Heterozygote variations in EDNRB gene were significantly enriched in three families and RET mutations were identified in one family. EDNRB variants showed an overall higher incidence and penetrance than RET in southern Chinese families cases.
Collapse
Affiliation(s)
- Hui-Yang Ding
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Wen Lei
- Maternal and Child Health Research Institute, Translational Medicine Center, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Shang-Jie Xiao
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Hua Deng
- Maternal and Child Health Research Institute, Translational Medicine Center, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Li-Ke Yuan
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Lu Xu
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Jia-Liang Zhou
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Rong Huang
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Yuan-Long Fang
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Qing-Yuan Wang
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Ying Zhang
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Liang Zhang
- Maternal and Child Health Research Institute, Translational Medicine Center, Guangdong Women and Children Hospital, Guangzhou, 511400, China.
| | - Xiao-Chun Zhu
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, 511400, China.
| |
Collapse
|
41
|
Basilisco G, Marchi M, Coletta M. Chronic intestinal pseudo-obstruction in adults: A practical guide to identify patient subgroups that are suitable for more specific treatments. Neurogastroenterol Motil 2024; 36:e14715. [PMID: 37994282 DOI: 10.1111/nmo.14715] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Chronic intestinal pseudo-obstruction is a rare and heterogeneous syndrome characterized by recurrent symptoms of intestinal obstruction with radiological features of dilated small or large intestine with air/fluid levels in the absence of any mechanical occlusive lesion. Several diseases may be associated with chronic intestinal pseudo-obstruction and in these cases, the prognosis and treatment are related to the underlying disease. Also, in its "primary or idiopathic" form, two subgroups of patients should be determined as they require a more specific therapeutic approach: patients whose chronic intestinal pseudo-obstruction is due to sporadic autoimmune/inflammatory mechanisms and patients whose neuromuscular changes are genetically determined. In a context of a widely heterogeneous adult population presenting chronic intestinal pseudo-obstruction, this review aims to summarize a practical diagnostic workup for identifying definite subgroups of patients who might benefit from more specific treatments, based on the etiology of their underlying condition.
Collapse
Affiliation(s)
- Guido Basilisco
- Gastroenterology and Endoscopic Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Margherita Marchi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Marina Coletta
- Gastroenterology and Endoscopic Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
42
|
Kanai SM, Clouthier DE. Endothelin signaling in development. Development 2023; 150:dev201786. [PMID: 38078652 PMCID: PMC10753589 DOI: 10.1242/dev.201786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Since the discovery of endothelin 1 (EDN1) in 1988, the role of endothelin ligands and their receptors in the regulation of blood pressure in normal and disease states has been extensively studied. However, endothelin signaling also plays crucial roles in the development of neural crest cell-derived tissues. Mechanisms of endothelin action during neural crest cell maturation have been deciphered using a variety of in vivo and in vitro approaches, with these studies elucidating the basis of human syndromes involving developmental differences resulting from altered endothelin signaling. In this Review, we describe the endothelin pathway and its functions during the development of neural crest-derived tissues. We also summarize how dysregulated endothelin signaling causes developmental differences and how this knowledge may lead to potential treatments for individuals with gene variants in the endothelin pathway.
Collapse
Affiliation(s)
- Stanley M. Kanai
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David E. Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
43
|
Gagnon H, Duguay S, Prasil P, Castilloux J. Short and Long-Term Outcomes in Hirschsprung Disease: Are the Syndrome-Associated Patients Really Doing Worse? J Pediatr Gastroenterol Nutr 2023; 77:592-596. [PMID: 37496148 DOI: 10.1097/mpg.0000000000003896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Given the lack of data to help caregivers in the follow-up of Hirschsprung disease (HD), this study aimed to compare the functional outcomes of isolated Hirschsprung disease (I-HD) to syndrome-associated Hirschsprung disease (SA-HD) at 1, 3, 5, and 10 years. METHODS A retrospective chart review of patients diagnosed with HD between January 1990 and May 2021 at our pediatric center was performed to collect data on patient characteristics, investigations, and treatments. Ninety-five patients were identified, of whom 76 were included in the study. SA-HD is defined as a syndrome known to be associated with HD or cognitive impairment. RESULTS Patient characteristics were comparable between groups ( P > 0.05). There were 52 patients with I-HD and 24 with SA-HD. The patients median age was 9 days at diagnosis and 1.5 month at surgery. SA-HD patients became bowel continent at a significantly older age (mean age 8.43 vs 4.94 years, P = 0.0471) and received more bowel continence medications. At 5 years, SA-HD patients requiring ≥2 medications for bowel continence represented 54.5% versus 11.1% of I-HD patients ( P = 0.009). Lastly, SA-HD patients had urinary incontinence at a significantly older age ( P = 0.0136, 5 years). CONCLUSION Clinicians should be aware that SA-HD patients are more prone to bladder dysfunction and became bowel continent at an older age than I-HD patients. They need more and prolonged bowel management medications, and other important complications need to be addressed in patient care. These results should prompt a longer follow-up period for these patients, especially in SA-HD.
Collapse
Affiliation(s)
- Hugo Gagnon
- From the Department of Pediatrics, Centre Mère-Enfant Soleil du Centre Hospitalier de l'Université Laval, Quebec, Canada
| | - Sophie Duguay
- From the Department of Pediatrics, Centre Mère-Enfant Soleil du Centre Hospitalier de l'Université Laval, Quebec, Canada
| | - Pascale Prasil
- From the Department of Pediatrics, Centre Mère-Enfant Soleil du Centre Hospitalier de l'Université Laval, Quebec, Canada
- Department of Surgery, Centre Mère-Enfant Soleil du Centre Hospitalier de l'Université Laval, Quebec, Canada
| | - Julie Castilloux
- From the Department of Pediatrics, Centre Mère-Enfant Soleil du Centre Hospitalier de l'Université Laval, Quebec, Canada
- the Department of Pediatric Gastro-Enterology, Centre Mère-Enfant Soleil du Centre Hospitalier de l'Université Laval, Quebec, Canada
| |
Collapse
|
44
|
Jacobs-Li J, Tang W, Li C, Bronner ME. Single-cell profiling coupled with lineage analysis reveals vagal and sacral neural crest contributions to the developing enteric nervous system. eLife 2023; 12:e79156. [PMID: 37877560 PMCID: PMC10627514 DOI: 10.7554/elife.79156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 10/23/2023] [Indexed: 10/26/2023] Open
Abstract
During development, much of the enteric nervous system (ENS) arises from the vagal neural crest that emerges from the caudal hindbrain and colonizes the entire gastrointestinal tract. However, a second ENS contribution comes from the sacral neural crest that arises in the caudal neural tube and populates the post-umbilical gut. By coupling single-cell transcriptomics with axial-level-specific lineage tracing in avian embryos, we compared the contributions of embryonic vagal and sacral neural crest cells to the chick ENS and the associated peripheral ganglia (Nerve of Remak and pelvic plexuses). At embryonic day (E) 10, the two neural crest populations form overlapping subsets of neuronal and glia cell types. Surprisingly, the post-umbilical vagal neural crest much more closely resembles the sacral neural crest than the pre-umbilical vagal neural crest. However, some differences in cluster types were noted between vagal and sacral derived cells. Notably, RNA trajectory analysis suggests that the vagal neural crest maintains a neuronal/glial progenitor pool, whereas this cluster is depleted in the E10 sacral neural crest which instead has numerous enteric glia. The present findings reveal sacral neural crest contributions to the hindgut and associated peripheral ganglia and highlight the potential influence of the local environment and/or developmental timing in differentiation of neural crest-derived cells in the developing ENS.
Collapse
Affiliation(s)
- Jessica Jacobs-Li
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Weiyi Tang
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Can Li
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
45
|
Montalva L, Cheng LS, Kapur R, Langer JC, Berrebi D, Kyrklund K, Pakarinen M, de Blaauw I, Bonnard A, Gosain A. Hirschsprung disease. Nat Rev Dis Primers 2023; 9:54. [PMID: 37828049 DOI: 10.1038/s41572-023-00465-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Hirschsprung disease (HSCR) is a rare congenital intestinal disease that occurs in 1 in 5,000 live births. HSCR is characterized by the absence of ganglion cells in the myenteric and submucosal plexuses of the intestine. Most patients present during the neonatal period with the first meconium passage delayed beyond 24 h, abdominal distension and vomiting. Syndromes associated with HSCR include trisomy 21, Mowat-Wilson syndrome, congenital central hypoventilation syndrome, Shah-Waardenburg syndrome and cartilage-hair hypoplasia. Multiple putative genes are involved in familial and isolated HSCR, of which the most common are the RET proto-oncogene and EDNRB. Diagnosis consists of visualization of a transition zone on contrast enema and confirmation via rectal biopsy. HSCR is typically managed by surgical removal of the aganglionic bowel and reconstruction of the intestinal tract by connecting the normally innervated bowel down to the anus while preserving normal sphincter function. Several procedures, namely Swenson, Soave and Duhamel procedures, can be undertaken and may include a laparoscopically assisted approach. Short-term and long-term comorbidities include persistent obstructive symptoms, enterocolitis and soiling. Continued research and innovation to better understand disease mechanisms holds promise for developing novel techniques for diagnosis and therapy, and improving outcomes in patients.
Collapse
Affiliation(s)
- Louise Montalva
- Department of Paediatric Surgery, Robert-Debré Children's University Hospital, Paris, France.
- Faculty of Health, Paris-Cité University, Paris, France.
- NeuroDiderot, INSERM UMR1141, Paris, France.
| | - Lily S Cheng
- Division of Paediatric Surgery, Texas Children's Hospital, Houston, TX, USA
- Division of Paediatric Surgery, University of Virginia, Charlottesville, VA, USA
| | - Raj Kapur
- Department of Pathology, Seattle Children's Hospital, Seattle, WA, USA
| | - Jacob C Langer
- Division of Paediatric Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dominique Berrebi
- Department of Pathology, Robert-Debré and Necker Children's University Hospital, Paris, France
| | - Kristiina Kyrklund
- Department of Paediatric Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Mikko Pakarinen
- Department of Paediatric Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Ivo de Blaauw
- Department of Surgery, Division of Paediatric Surgery, Radboudumc-Amalia Children's Hospital, Nijmegen, Netherlands
| | - Arnaud Bonnard
- Department of Paediatric Surgery, Robert-Debré Children's University Hospital, Paris, France
- Faculty of Health, Paris-Cité University, Paris, France
- NeuroDiderot, INSERM UMR1141, Paris, France
| | - Ankush Gosain
- Department of Paediatric Surgery, Children's Hospital Colorado, Aurora, CO, USA.
| |
Collapse
|
46
|
Yip JLK, Xavier S, Balasuriya GK, Hill-Yardin EL, Spencer SJ. Macrophage regulation of the "second brain": CD163 intestinal macrophages interact with inhibitory interneurons to regulate colonic motility - evidence from the Cx3cr1-Dtr rat model. Front Immunol 2023; 14:1269890. [PMID: 37868978 PMCID: PMC10585175 DOI: 10.3389/fimmu.2023.1269890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Intestinal macrophages are well-studied for their conventional roles in the immune response against pathogens and protecting the gut from chronic inflammation. However, these macrophages may also have additional functional roles in gastrointestinal motility under typical conditions. This is likely to occur via both direct and indirect influences on gastrointestinal motility through interaction with myenteric neurons that contribute to the gut-brain axis, but this mechanism is yet to be properly characterised. The CX3CR1 chemokine receptor is expressed in the majority of intestinal macrophages, so we used a conditional knockout Cx3cr1-Dtr (diphtheria toxin receptor) rat model to transiently ablate these cells. We then utilized ex vivo video imaging to evaluate colonic motility. Our previous studies in brain suggested that Cx3cr1-expressing cells repopulate by 7 days after depletion in this model, so we performed our experiments at both the 48 hr (macrophage depletion) and 7-day (macrophage repopulation) time points. We also investigated whether inhibitory neuronal input driven by nitric oxide from the enteric nervous system is required for the regulation of colonic motility by intestinal macrophages. Our results demonstrated that CD163-positive resident intestinal macrophages are important in regulating colonic motility in the absence of this major inhibitory neuronal input. In addition, we show that intestinal macrophages are indispensable in maintaining a healthy intestinal structure. Our study provides a novel understanding of the interplay between the enteric nervous system and intestinal macrophages in colonic motility. We highlight intestinal macrophages as a potential therapeutic target for gastrointestinal motility disorders when inhibitory neuronal input is suppressed.
Collapse
Affiliation(s)
- Jackson L. K. Yip
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Soniya Xavier
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Gayathri K. Balasuriya
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
| | - Elisa L. Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Sarah J. Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Yang W, Chen SC, Wang TE, Tsai PS, Chen JC, Chen PL. L1cam alternative shorter transcripts encoding the extracellular domains were overexpressed in the intestine of L1cam knockdown mice. Gene 2023; 881:147643. [PMID: 37453721 DOI: 10.1016/j.gene.2023.147643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/25/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Hirschsprung disease (HSCR) is a congenital disorder of functional bowel obstruction due to the absence of enteric ganglia in distal bowel. Different L1cam variants were reportedly associated with L1cam syndrome and HSCR, whose phenotypes lacked predictable relevance to their genotypes. Using next-generation sequencing (NGS), we found an L1CAM de novo frameshift mutation in a female with mild hydrocephalus and skip-type HSCR. A nearly identical L1cam variant was introduced into FVB/NJ mice via the CRISPR-EZ method. A silent mutation was created via ssODN to gain an artificial Ncol restriction enzyme site for easier genotyping. Six L1cam protein-coding alternative transcripts were quantitatively measured. Immunofluorescence staining with polyclonal and monoclonal L1cam antibodies was used to characterize L1cam isoform proteins in enteric ganglia. Fifteen mice, seven males and eight females, generated via CRISPR-EZ, were confirmed to carry the L1cam frameshift variant, resulting in a premature stop codon. There was no prominent hydrocephalus nor HSCR-like presentation in these mice, but male infertility was noticed after observation for three generations in a total of 176 mice. Full-length L1cam transcripts were detected at a very low level in the intestinal tissues and almost none in the brain of these mice. Alternative shorter transcripts encoding the extracellular domains were overexpressed in the intestine of L1cam knockdown mice. Immunofluorescence confirmed no fulllength L1cam protein in enteric ganglia. These shorter L1cam isoform proteins might play a role in protecting L1cam knockdown mice from HSCR.
Collapse
Affiliation(s)
- Wendy Yang
- Department of Surgery, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Pediatric Research Center, Chang Gung Children's Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Szu-Chieh Chen
- Pediatric Research Center, Chang Gung Children's Hospital, Taoyuan, Taiwan
| | - Tse-En Wang
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 10617 Taipei, Taiwan
| | - Pei-Shiue Tsai
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, 10617 Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, 10617 Taipei, Taiwan
| | - Jeng-Chang Chen
- Department of Surgery, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Pediatric Research Center, Chang Gung Children's Hospital, Taoyuan, Taiwan.
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taiwan; Departments of Medical Genetics, National Taiwan University Hospital, Taiwan; Departments of Internal Medicine, National Taiwan University Hospital, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
48
|
Laddach A, Chng SH, Lasrado R, Progatzky F, Shapiro M, Erickson A, Sampedro Castaneda M, Artemov AV, Bon-Frauches AC, Amaniti EM, Kleinjung J, Boeing S, Ultanir S, Adameyko I, Pachnis V. A branching model of lineage differentiation underpinning the neurogenic potential of enteric glia. Nat Commun 2023; 14:5904. [PMID: 37737269 PMCID: PMC10516949 DOI: 10.1038/s41467-023-41492-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
Glial cells have been proposed as a source of neural progenitors, but the mechanisms underpinning the neurogenic potential of adult glia are not known. Using single cell transcriptomic profiling, we show that enteric glial cells represent a cell state attained by autonomic neural crest cells as they transition along a linear differentiation trajectory that allows them to retain neurogenic potential while acquiring mature glial functions. Key neurogenic loci in early enteric nervous system progenitors remain in open chromatin configuration in mature enteric glia, thus facilitating neuronal differentiation under appropriate conditions. Molecular profiling and gene targeting of enteric glial cells in a cell culture model of enteric neurogenesis and a gut injury model demonstrate that neuronal differentiation of glia is driven by transcriptional programs employed in vivo by early progenitors. Our work provides mechanistic insight into the regulatory landscape underpinning the development of intestinal neural circuits and generates a platform for advancing glial cells as therapeutic agents for the treatment of neural deficits.
Collapse
Affiliation(s)
- Anna Laddach
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Song Hui Chng
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Experimental Drug Development Centre A*STAR 10 Biopolis Road, Chromos, 138670, Singapore
| | - Reena Lasrado
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- COMPASS Pathways PLC, Fora, 33 Broadwick St, London, W1F 0DQ, UK
| | - Fränze Progatzky
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Michael Shapiro
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Alek Erickson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Marisol Sampedro Castaneda
- Kinases and Brain Development Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Artem V Artemov
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Bienna, 1090, Austria
- Boehringer Ingelheim RCV, Vienna, Austria
| | - Ana Carina Bon-Frauches
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Eleni-Maria Amaniti
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Sainsbury Wellcome Centre, London, UK
| | - Jens Kleinjung
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, CB21 6DG, UK
| | - Stefan Boeing
- Bioinformatics and Biostatistics Science Technology Platform, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Sila Ultanir
- Kinases and Brain Development Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17165, Sweden
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Bienna, 1090, Austria
| | - Vassilis Pachnis
- Nervous System Development and Homeostasis Laboratory, the Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
49
|
Eeftinck Schattenkerk LD, Musters GD, de Jonge WJ, van Heurn L(E, Derikx JPM. Should primary anastomosis be considered more? A retrospective analysis of anastomotic complications in young children. WORLD JOURNAL OF PEDIATRIC SURGERY 2023; 6:e000565. [PMID: 37671119 PMCID: PMC10476123 DOI: 10.1136/wjps-2023-000565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/26/2023] [Indexed: 09/07/2023] Open
Abstract
Objective Little is known about intestinal anastomotic leakage and stenosis in young children (≤3 years of age). The purpose of this study is to answer the following questions: (1) what is the incidence of anastomotic stenosis and leakage in infants? (2) which surgical diseases entail the highest incidence of anastomotic stenosis and leakage? (3) what are perioperative factors associated with anastomotic stenosis and leakage? Methods Patients who underwent an intestinal anastomosis during primary abdominal surgery in our tertiary referral centre between 1998 and 2018 were retrospectively included. Both general incidence and incidence per disease of anastomotic complications were determined. Technical risk factors (location and type of anastomosis, mode of suturing, and suture resorption time) were evaluated by multivariate Cox regression for anastomotic stenosis. Gender and American Society of Anaesthesiology (ASA) score of ≥III were evaluated by χ2 test for anastomotic leakage. Results In total, 477 patients underwent an anastomosis. The most prominent diseases are intestinal atresia (30%), Hirschsprung's disease (29%), and necrotizing enterocolitis (14%). Anastomotic stenosis developed in 7% (34/468) of the patients with highest occurrence in necrotizing enterocolitis (14%, 9/65). Colonic anastomosis was associated with an increased risk of anastomotic stenosis (hazard ratio (HR) =3.6, 95% CI 1.8 to 7.5). No technical features (type of anastomosis, suture resorption time and mode of suturing) were significantly associated with stenosis development. Anastomotic leakage developed in 5% (22/477) of the patients, with the highest occurrence in patients with intestinal atresia (6%, 9/143). An ASA score of ≥III (p=0.03) and male gender (p=0.03) were significantly associated with anastomotic leakage. Conclusions Both anastomotic stenosis and leakage are major surgical complications. Identifying more patient specific factors can result in better treatment selection, which should not solely be based on the type of disease.
Collapse
Affiliation(s)
- Laurens D. Eeftinck Schattenkerk
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Gijsbert D. Musters
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of General, Visceral, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - L.W. (Ernest) van Heurn
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joep PM Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Yang Y, Xia L, Yang W, Wang Z, Meng W, Zhang M, Ma Q, Gou J, Wang J, Shu Y, Wu X. Transcriptome profiling of intact bowel wall reveals that PDE1A and SEMA3D are possible markers with roles in enteric smooth muscle apoptosis, proliferative disorders, and dysautonomia in Crohn's disease. Front Genet 2023; 14:1194882. [PMID: 37727374 PMCID: PMC10505932 DOI: 10.3389/fgene.2023.1194882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023] Open
Abstract
Background: Inflammatory bowel disease (IBD) is a complex and multifactorial inflammatory condition, comprising Crohn's disease (CD) and ulcerative colitis (UC). While numerous studies have explored the immune response in IBD through transcriptional profiling of the enteric mucosa, the subtle distinctions in the pathogenesis of Crohn's disease and ulcerative colitis remain insufficiently understood. Methods: The intact bowel wall specimens from IBD surgical patients were divided based on their inflammatory status into inflamed Crohn's disease (iCD), inflamed ulcerative colitis (iUC) and non-inflamed (niBD) groups for RNA sequencing. Differential mRNA GO (Gene Ontology), and KEGG (Kyoto Encyclopedia of Genes and Genomes), and GSEA (Gene Set Enrichment Analysis) bioinformatic analyses were performed with a focus on the enteric autonomic nervous system (ANS) and smooth muscle cell (SMC). The transcriptome results were validated by quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC). Results: A total of 2099 differentially expressed genes were identified from the comparison between iCD and iUC. Regulation of SMC apoptosis and proliferation were significantly enriched in iCD, but not in iUC. The involved gene PDE1A in iCD was 4-fold and 1.5-fold upregulated at qPCR and IHC compared to that in iUC. Moreover, only iCD was significantly associated with the gene sets of ANS abnormality. The involved gene SEMA3D in iCD was upregulated 8- and 5-fold at qPCR and IHC levels compared to iUC. Conclusion: These findings suggest that PDE1A and SEMA3D may serve as potential markers implicated in enteric smooth muscle apoptosis, proliferative disorders, and dysautonomia specifically in Crohn's disease.
Collapse
Affiliation(s)
- Yun Yang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of General Surgery, West China Chengdu Shangjin Nanfu Hospital, Sichuan University, Chengdu, China
| | - Lin Xia
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenming Yang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenjian Meng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Mingming Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Department of General Surgery, West China Chengdu Shangjin Nanfu Hospital, Sichuan University, Chengdu, China
| | - Qin Ma
- Department of General Surgery, West China Chengdu Shangjin Nanfu Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Junhe Gou
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Junjian Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Shu
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Wu
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Colorectal and Pelvic Floor Center, West China Tianfu Hospital, Sichuan University, Chengdu, China
| |
Collapse
|